1
|
Victor Atoki A, Aja PM, Shinkafi TS, Ondari EN, Adeniyi AI, Fasogbon IV, Dangana RS, Shehu UU, Akin-Adewumi A. Exploring the versatility of Drosophila melanogaster as a model organism in biomedical research: a comprehensive review. Fly (Austin) 2025; 19:2420453. [PMID: 39722550 DOI: 10.1080/19336934.2024.2420453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 10/16/2024] [Accepted: 10/16/2024] [Indexed: 12/28/2024] Open
Abstract
Drosophila melanogaster is a highly versatile model organism that has profoundly advanced our understanding of human diseases. With more than 60% of its genes having human homologs, Drosophila provides an invaluable system for modelling a wide range of pathologies, including neurodegenerative disorders, cancer, metabolic diseases, as well as cardiac and muscular conditions. This review highlights key developments in utilizing Drosophila for disease modelling, emphasizing the genetic tools that have transformed research in this field. Technologies such as the GAL4/UAS system, RNA interference (RNAi) and CRISPR-Cas9 have enabled precise genetic manipulation, with CRISPR-Cas9 allowing for the introduction of human disease mutations into orthologous Drosophila genes. These approaches have yielded critical insights into disease mechanisms, identified novel therapeutic targets and facilitated both drug screening and toxicological studies. Articles were selected based on their relevance, impact and contribution to the field, with a particular focus on studies offering innovative perspectives on disease mechanisms or therapeutic strategies. Our findings emphasize the central role of Drosophila in studying complex human diseases, underscoring its genetic similarities to humans and its effectiveness in modelling conditions such as Alzheimer's disease, Parkinson's disease and cancer. This review reaffirms Drosophila's critical role as a model organism, highlighting its potential to drive future research and therapeutic advancements.
Collapse
Affiliation(s)
| | - Patrick Maduabuchi Aja
- Department of Biochemistry, Kampala International University, Ishaka, Uganda
- Department of Biochemistry, Faculty of Science, Ebonyi State University, Abakaliki, Nigeria
| | | | - Erick Nyakundi Ondari
- Department of Biochemistry, Kampala International University, Ishaka, Uganda
- School of Pure and Applied Sciences, Department of Biological Sciences, Kisii University, Kisii, Kenya
| | | | | | | | - Umar Uthman Shehu
- Department of Physiology, Kampala International University, Ishaka, Uganda
| | | |
Collapse
|
2
|
Galushkin A, Gozes I. Intranasal NAP (Davunetide): Neuroprotection and circadian rhythmicity. Adv Drug Deliv Rev 2025; 220:115573. [PMID: 40185278 DOI: 10.1016/j.addr.2025.115573] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Revised: 02/05/2025] [Accepted: 03/24/2025] [Indexed: 04/07/2025]
Abstract
In this review we examine the neuroprotective potential of NAP (davunetide), a small peptide derived from Activity-Dependent Neuroprotective Protein (ADNP), in the context of neurodevelopmental and neurodegenerative disorders. ADNP, a protein essential for brain development and function, is associated with tauopathy-related diseases, such as Alzheimer's Disease (AD), and circadian rhythm regulation. NAP enhances microtubule stability and prevents tauopathy. In preclinical studies, NAP shows promise in improving cognitive performance and correcting behavioral deficits in different models. Clinical studies on NAP (davunetide) administered via intranasal delivery have demonstrated its safety, favorable bioavailability, and potential efficacy in improving cognitive function, making it a viable therapeutic option. In the pure tauopathy, progressive supranuclear palsy, NAP (davunetide) significantly slowed disease progression in women in a phase II-III clinical trial. Additionally, the complex interactions between ADNP, associated pathways, and circadian regulation and the extensive NAP compensation upon ADNP deficiency attest to further clinical development. Thus, NAP is an example of a reductionist approach in drug delivery, replacing/enhancing the critical large ADNP-related pathways including dysregulated microtubules and tauopathy with a small brain bioavailable investigational drug, davunetide.
Collapse
Affiliation(s)
- Artur Galushkin
- Dr. Diana and Zelman Elton Laboratory for Molecular Neuroendocrinology, Department of Human Molecular Genetics and Biochemistry, Faculty of Medical & Health Sciences, Adams Super Center for Brain Studies and Sagol School of Neuroscience, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Illana Gozes
- Dr. Diana and Zelman Elton Laboratory for Molecular Neuroendocrinology, Department of Human Molecular Genetics and Biochemistry, Faculty of Medical & Health Sciences, Adams Super Center for Brain Studies and Sagol School of Neuroscience, Tel Aviv University, Tel Aviv 6997801, Israel.
| |
Collapse
|
3
|
Ding H, Madan S, Searls E, McNulty M, Low S, Li Z, Ho K, Rahman S, Igwe A, Popp Z, Hwang PH, De Anda-Duran I, Kolachalama VB, Mez J, Alosco ML, Thomas RJ, Au R, Lin H. Exploring nightly variability and clinical influences on sleep measures: insights from a digital brain health platform. Sleep Med 2025; 131:106532. [PMID: 40306226 DOI: 10.1016/j.sleep.2025.106532] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 03/24/2025] [Accepted: 04/21/2025] [Indexed: 05/02/2025]
Abstract
BACKGROUND Digital technology offers a convenient way to continuously monitor sleep and assess night-to-night variability, particularly in aging populations where traditional self-reported sleep assessments may be limited. AIMS This study aimed to investigate nightly variability in sleep measures obtained via a ring oximeter sensor in older adults and to explore the influence of demographic and cognitive factors on the stability of these metrics. METHODS The study included 62 participants (mean age 74, 67.7 % women, 90.3 % White) from the Boston University Alzheimer's Disease Research Center (BU ADRC) cohort. Each participant wore a SleepImage Ring for at least three consecutive nights. Thirty-four continuous sleep measures, such as mean SpO2 and apnea-hypopnea index within unstable sleep, were analyzed. Night-to-night variability was assessed using intraclass correlation coefficients (ICC) based on a two-way random-effects model. Subgroup analyses examined variability by sex, age, and cognitive status. Group-level changes were assessed using one-way repeated measures ANOVA. RESULTS Seven sleep measures demonstrated high stability across nights (ICC: 0.70-0.88), with average heart rate being the most stable, followed by mean SpO2 and apnea-hypopnea indices. Sleep latency exhibited the highest variability. Stability improved between the second and third nights compared to the first and second nights. Women and participants under 75 years old showed greater stability in several metrics, while cognitively intact individuals exhibited more consistent breathing-related measures. CONCLUSION At least three nights of monitoring are required for reliable estimates of key sleep metrics. Expanding studies with larger samples and extended monitoring periods could further elucidate sleep variability as a potential non-invasive marker for general health.
Collapse
Affiliation(s)
- Huitong Ding
- Department of Anatomy and Neurobiology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA; The Framingham Heart Study, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Sanskruti Madan
- Department of Anatomy and Neurobiology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Edward Searls
- Department of Anatomy and Neurobiology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Matthew McNulty
- Department of Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Spencer Low
- Department of Anatomy and Neurobiology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Zexu Li
- Department of Anatomy and Neurobiology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Kristi Ho
- Department of Anatomy and Neurobiology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Salman Rahman
- Department of Anatomy and Neurobiology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Akwaugo Igwe
- Department of Anatomy and Neurobiology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Zachary Popp
- Department of Anatomy and Neurobiology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Phillip H Hwang
- Department of Anatomy and Neurobiology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA; The Framingham Heart Study, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA; Department of Epidemiology, Boston University School of Public Health, Boston, MA, USA
| | - Ileana De Anda-Duran
- School of Public Health and Tropical Medicine, Tulane University, New Orleans, LA, USA
| | - Vijaya B Kolachalama
- Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA; Department of Computer Science and Faculty of Computing & Data Sciences, Boston University, Boston, MA, USA
| | - Jesse Mez
- Department of Anatomy and Neurobiology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA; Boston University Alzheimer's Disease Research Center, Boston University CTE Center, Departments of Neurology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA; Department of Neurology, Boston Medical Center, USA
| | - Michael L Alosco
- Department of Anatomy and Neurobiology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA; Boston University Alzheimer's Disease Research Center, Boston University CTE Center, Departments of Neurology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA; Department of Neurology, Boston Medical Center, USA
| | - Robert J Thomas
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Rhoda Au
- Department of Anatomy and Neurobiology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA; The Framingham Heart Study, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA; Department of Epidemiology, Boston University School of Public Health, Boston, MA, USA; Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA; Boston University Alzheimer's Disease Research Center, Boston University CTE Center, Departments of Neurology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA; Slone Epidemiology Center, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Honghuang Lin
- Department of Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA.
| |
Collapse
|
4
|
Jiao H, Huang S, Cheng W, Feng J, Yu J. Associations of physical activity, sedentary behavior, and sleep with risk of incident Parkinson's disease: A prospective cohort study of 401,697 participants. Chin Med J (Engl) 2025; 138:819-828. [PMID: 39967299 PMCID: PMC11970816 DOI: 10.1097/cm9.0000000000003399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Indexed: 02/20/2025] Open
Abstract
BACKGROUND Physical activity, sedentary behavior (SB), and sleep duration are associated with brain health. Effects of those on developing Parkinson's disease (PD) are poorly investigated. This study aimed to examine the independent and joint associations of physical activity, SB, sleep with PD risk. METHODS We analyzed data on 401,697 participants from the UK Biobank cohort, which was enrolled in 2006-2010. Physical activities were measured based on a questionnaire. Sleep and SB time were defined through self-reported total number of hours. Models fitted with restricted cubic spline were conducted to test for linear and non-linear shapes of each association. Cox proportional hazards regression models were used to estimate the association of three modifiable behaviors. RESULTS Our analytic sample included 401,697 participants with 3030 identified cases of PD (mean age, 63 years; 62.9% male). PD risk was 18% lower in the high total physical activity group (95% CI, 0.75-0.90), 22% lower in the high leisure-time physical activity (LTPA) group (95% CI, 0.71-0.86) compared with the low level and 14% higher in the high sleep duration group (95% CI, 1.05-1.24) compared to moderate group. Total SB time was irrelevant with PD risk, while high TV viewing showed a 12% increase of PD risk compared to the low group (95% CI, 1.02-1.22). Low computer use (0 h/day) was associated with a 14% higher risk compared to 1 h/day use (95% CI, 1.04-1.26). Those associations were independent. A combination of 7 h/day sleep, moderate-to-high computer use, and moderate-to-vigorous intensity of LTPA showed lowest PD risk (HR, 0.70; 95% CI, 0.57-0.85). CONCLUSIONS Physical activity, SB, and sleep were associated with PD risks separately. Our findings emphasize the possibility for changing these three daily activities concurrently to lower the risk of PD. These findings may promote an active lifestyle for PD prevention.
Collapse
Affiliation(s)
- Haishan Jiao
- Department of Neurology and Institute of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai 200040, China
| | - Shuyi Huang
- Department of Neurology and Institute of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai 200040, China
| | - Wei Cheng
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai 200433, China
- Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence, Fudan University, Ministry of Education, Shanghai 200433, China
- Fudan ISTBI-ZJNU Algorithm Centre for Brain-Inspired Intelligence, Zhejiang Normal University, Jinhua, Zhejiang 321004, China
- Shanghai Medical College and Zhongshan Hospital Immunotherapy Technology Transfer Center, Shanghai 200032, China
| | - Jianfeng Feng
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai 200433, China
- Key Laboratory of Computational Neuroscience and Brain-Inspired Intelligence, Fudan University, Ministry of Education, Shanghai 200433, China
| | - Jintai Yu
- Department of Neurology and Institute of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai 200040, China
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai 200433, China
| |
Collapse
|
5
|
Zhu W, Hu Y, Shi Y, Bao H, Cheng X, Jiang M, Peng Z, Song J, Fang F, Jian C, Yuan W, Chen J, Shu X. Sleep deprivation accelerates Parkinson's disease via modulating gut microbiota associated microglial activation and oxidative stress. Microbiol Res 2025; 293:128077. [PMID: 39889629 DOI: 10.1016/j.micres.2025.128077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2024] [Revised: 12/01/2024] [Accepted: 01/19/2025] [Indexed: 02/03/2025]
Abstract
The interplay between Parkinson's disease (PD) and sleep disturbances suggests that sleep problems constitute a risk factor for PD progression, but the underlying mechanisms remain unclear. Microglial activation and oxidative stress are considered to play an important role in the pathogenesis of aging and neurodegenerative diseases. We hypothesized that sleep deprivation (SD) could exacerbate PD progression via modulating microglial activation and oxidative stress. To test this hypothesis, we established a PD mouse model using 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP), then subjected the mice to SD. A battery of behavioral tests, including rotarod, pole, adhesive removal, and open field tests, were used to assess motor function. Our study showed that SD exacerbated motor deficits, loss of tyrosine hydroxylase (TH), microglial activation and oxidative stress damage in PD model mice. Fecal microbiota transplantation experiments revealed that SD mediated PD progression, microglial activation and oxidative stress via the gut microbiota. 16S rRNA sequencing analysis indicated that SD increased the abundances of bacteria such as Bacteroidaceae, while decreasing the abundances of bacteria including Lactobacillus. Non-targeted metabolomic analysis of gut microbiota-derived metabolites revealed that SD significantly increased the production of adenosine (ADO), a purine metabolite. Probiotic supplementation reversed the effects of SD on motor deficits, dopaminergic neuron loss, microglial activation and oxidative stress damage in PD mice; it also decreased SD-induced ADO production. Administration of Adenosine A2A receptor (A2AR) inhibitors, Istradefylline (Ist), attenuated the roles of SD and ADO in promoting microglial activation, oxidative stress and PD progression. Taken together, our findings indicate that SD accelerates PD progression via regulating microbiota associated microglial activation and oxidative stress, suggesting that efforts to improve sleep quality can be used to prevent and treat PD.
Collapse
Affiliation(s)
- Wenzhong Zhu
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, PR China
| | - Yuan Hu
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, PR China
| | - Yongping Shi
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, PR China
| | - Haijun Bao
- Department of Emergency Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Jiefang Road No,1277, Wuhan, Hubei 430022, China
| | - Xukai Cheng
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, PR China
| | - Mi Jiang
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, PR China
| | - Zuojie Peng
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, PR China
| | - Jia Song
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, PR China
| | - Feifei Fang
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, PR China
| | - Chenxing Jian
- Department of Colorectal Surgery, Affiliated Hospital of Putian University, Putian, Fujian 351100, China
| | - Wenzheng Yuan
- Department of Gastrointestinal Surgery II, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
| | - Jinghuang Chen
- Department of Emergency Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Jiefang Road No,1277, Wuhan, Hubei 430022, China.
| | - Xiaogang Shu
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, PR China.
| |
Collapse
|
6
|
Yang MY, Wu CN, Lin YT, Tsai MH, Hwang CF, Yang CH. Dissecting the Circadian Clock and Toll-like Receptor Gene Alterations in Meniere's Disease and Vestibular Migraine. Otolaryngol Head Neck Surg 2025; 172:999-1005. [PMID: 39675041 DOI: 10.1002/ohn.1085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Revised: 11/03/2024] [Accepted: 11/22/2024] [Indexed: 12/17/2024]
Abstract
OBJECTIVE To investigate alterations in the expression of circadian clock and Toll-like receptor (TLR) genes in peripheral blood (PB) leukocytes of patients with Meniere's disease (MD) and vestibular migraine (VM), and determine whether these gene expressions can differentiate MD from VM. STUDY DESIGN Observational prospective study. SETTING Tertiary academic medical center. METHODS PB leukocytes were collected from patients diagnosed with MD and VM during recent vertigo attacks, as well as from healthy controls. The expression levels of 9 circadian clock genes and 6 TLR genes were analyzed using real-time quantitative reverse transcriptase-polymerase chain reaction. RESULTS Sixty-nine participants were enrolled, including 28 patients with MD, 14 patients with VM, and 27 healthy controls. Both MD and VM groups showed lower expression of PER1 compared to the control group (P < .01). The VM group exhibited significantly lower expression of PER1, PER2, CRY1, BMAL1, CLOCK, and TIM compared to the MD group (all P < .001). The MD group had higher TLR9 expression than the control group, and elevated TLR4, TLR8, and TLR9 expression compared to the VM group (P < .05). In the VM group, patients with severe dizziness handicaps had significantly lower expression of PER2, CRY1, CRY2, and CK1ε compared to those with mild to moderate handicaps (P < .05). CONCLUSION This study identifies distinct alterations in the circadian clock and TLR gene expression in MD and VM, suggesting potential differences in the pathogenesis of these 2 vertiginous disorders and highlighting the possibility of these gene expressions as biomarkers for differentiation.
Collapse
Affiliation(s)
- Ming-Yu Yang
- Department of Otolaryngology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
- Graduate Institute of Clinical Medical Sciences, College of Medicine, Chang Gung University, Tao-Yuan, Taiwan
| | - Ching-Nung Wu
- Department of Otolaryngology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Yu-Tsai Lin
- Department of Otolaryngology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
- Graduate Institute of Clinical Medical Sciences, College of Medicine, Chang Gung University, Tao-Yuan, Taiwan
- School of Medicine, College of Medicine, National Sun Yat-sen University, Kaohsiung, Taiwan
| | - Ming-Hsien Tsai
- Department of Otolaryngology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
- Graduate Institute of Clinical Medical Sciences, College of Medicine, Chang Gung University, Tao-Yuan, Taiwan
| | - Chung-Feng Hwang
- Department of Otolaryngology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
- School of Medicine, College of Medicine, National Sun Yat-sen University, Kaohsiung, Taiwan
| | - Chao-Hui Yang
- Department of Otolaryngology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
- School of Medicine, College of Medicine, National Sun Yat-sen University, Kaohsiung, Taiwan
- School of Medicine, Chang Gung University College of Medicine, Taoyuan, Taiwan
| |
Collapse
|
7
|
Fanciulli A, Sixel-Döring F, Buhmann C, Krismer F, Hermann W, Winkler C, Woitalla D, Jost WH, Trenkwalder C, Höglinger G. Diagnosis and treatment of autonomic failure, pain and sleep disturbances in Parkinson's disease: guideline "Parkinson's disease" of the German Society of Neurology. J Neurol 2025; 272:90. [PMID: 39751950 PMCID: PMC11698777 DOI: 10.1007/s00415-024-12730-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 11/15/2024] [Accepted: 11/17/2024] [Indexed: 01/04/2025]
Abstract
BACKGROUND AND OBJECTIVE Non-motor symptoms frequently develop throughout the disease course of Parkinson's disease (PD), and pose affected individuals at risk of complications, more rapid disease progression and poorer quality of life. Addressing such symptom burden, the 2023 revised "Parkinson's disease" guideline of the German Society of Neurology aimed at providing evidence-based recommendations for managing PD non-motor symptoms, including autonomic failure, pain and sleep disturbances. METHODS Key PICO (Patient, Intervention, Comparison, Outcome) questions were formulated by the steering committee and refined by the assigned authors. Recommendations were drafted based on relevant studies, systematic reviews, meta-analyses and high-quality guidelines identified by the literature search. They were subsequently reviewed, revised, and voted by the Guideline Group in online consensus conferences. Consensus was achieved in case of > 75% agreement among the group members. The consensus was considered strong, if agreement was > 95%. RESULTS The guideline entails: (i) 10 PICOs and 23 recommendations on the diagnosis and treatment of urogenital, cardiovascular and gastrointestinal autonomic failure; (ii) four PICOs and four recommendations on the possible types of pain in PD individuals, their diagnosis and treatment; (iii) 11 PICOs and 11 recommendations on the screening, diagnosis and treatment of sleep disturbances and excessive daytime sleepiness in PD individuals, as well as on their prognostic implications. Thirty-one out of 38 recommendations achieved a strong consensus. CONCLUSION The current German PD guideline provides a practice-oriented and etiology-driven stepwise approach to the diagnosis and treatment of autonomic failure, pain and sleep disturbances in PD individuals.
Collapse
Affiliation(s)
- Alessandra Fanciulli
- Department of Neurology, Innsbruck Medical University, Anichstrasse 35, 6020, Innsbruck, Austria.
| | - Friederike Sixel-Döring
- Paracelsus-Elena-Klinik, Kassel, Germany
- Department of Neurology, Philipps-Universität, Marburg, Germany
| | - Carsten Buhmann
- Department of Neurology, University Clinic Eppendorf, Hamburg, Germany
| | - Florian Krismer
- Department of Neurology, Innsbruck Medical University, Anichstrasse 35, 6020, Innsbruck, Austria
| | - Wiebke Hermann
- Department of Neurology, Rostock University Medical Center, Rostock, Germany
- Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE) Rostock/Greifswald, Rostock, Germany
| | - Christian Winkler
- Department of Neurology, Lindenbrunn Hospital, Coppenbrügge, Germany
| | | | | | - Claudia Trenkwalder
- Paracelsus-Elena-Klinik, Kassel, Germany
- Department of Neurosurgery, University Medical Center Göttingen, Göttingen, Germany
| | - Günter Höglinger
- Department of Neurology, LMU University Hospital, Ludwig-Maximilians-Universität (LMU) München, Munich, Germany
- Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| |
Collapse
|
8
|
Castro-Pascual IC, Ferramola ML, Altamirano FG, Cargnelutti E, Devia CM, Delgado SM, Lacoste MG, Anzulovich AC. Circadian organization of clock factors, antioxidant defenses, and cognitive genes expression, is lost in the cerebellum of aged rats. Possible targets of therapeutic strategies for the treatment of age-related cerebellar disorders. Brain Res 2024; 1845:149195. [PMID: 39182901 DOI: 10.1016/j.brainres.2024.149195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 08/10/2024] [Accepted: 08/21/2024] [Indexed: 08/27/2024]
Abstract
Aging is a major risk factor for cognitive deficits, impaired locomotion, and gait disorders. Although oxidative stress and circadian disruption are involved in both normal aging and the pathogenesis of age-associated diseases, just a very few studies explore the consequences of aging on circadian rhythms in the cerebellum. Here, we investigated age-dependent changes in the circadian organization of the molecular clock, antioxidant defenses and synaptic plasticity-related factors, in the rat cerebellum, and discussed the impact of that altered temporal organization on the cognitive function of this brain area. Particularly, we examined the circadian patterns of Brain and muscle ARNT-like 1 (BMAL1) protein levels, Glutathione peroxidase 4 (GPx4) gene expression, GPx and Catalase (CAT) enzymes activity, reduced glutathione (GSH) levels, and the Brain-derived neurotrophic factor (Bdnf) and its Tyrosine kinase receptor B (TrkB) circadian expression. Endogenously-driven circadian rhythms of BMAL1, GPx4, CAT, GSH, and Bdnf/TrkB factors, were observed in the young rat cerebellum. The rhythms' acrophases show a circadian organization that might be crucial for the daily cerebellar-dependent cognitive functions. Notably, aging disrupted circadian rhythms and the temporal organization of BMAL1, antioxidant defenses, and cognitive Bdnf/TrkB gene expression. Increased oxidative stress and disruption of clock-controlled rhythms during aging, might precede and cause the loss of circadian organization in the aged cerebellum. We expect our results highlight circadian rhythms of the studied factors as new targets for the treatment of age-dependent cerebellar disorders.
Collapse
Affiliation(s)
| | - Mariana L Ferramola
- Department of Biochemistry, Faculty of Chemistry, Biochemistry and Pharmacy, National University of San Luis (UNSL), San Luis, Argentina; Laboratory of Chronobiology, IMIBIO-SL (CONICET-UNSL), Argentina.
| | | | - Ethelina Cargnelutti
- Department of Biochemistry, Faculty of Chemistry, Biochemistry and Pharmacy, National University of San Luis (UNSL), San Luis, Argentina; Laboratory of Chronobiology, IMIBIO-SL (CONICET-UNSL), Argentina
| | - Cristina M Devia
- Department of Biochemistry, Faculty of Chemistry, Biochemistry and Pharmacy, National University of San Luis (UNSL), San Luis, Argentina
| | - Silvia M Delgado
- Department of Biochemistry, Faculty of Chemistry, Biochemistry and Pharmacy, National University of San Luis (UNSL), San Luis, Argentina; Laboratory of Chronobiology, IMIBIO-SL (CONICET-UNSL), Argentina
| | - María G Lacoste
- Department of Biochemistry, Faculty of Chemistry, Biochemistry and Pharmacy, National University of San Luis (UNSL), San Luis, Argentina; Laboratory of Chronobiology, IMIBIO-SL (CONICET-UNSL), Argentina
| | - Ana C Anzulovich
- Department of Biochemistry, Faculty of Chemistry, Biochemistry and Pharmacy, National University of San Luis (UNSL), San Luis, Argentina; Laboratory of Chronobiology, IMIBIO-SL (CONICET-UNSL), Argentina.
| |
Collapse
|
9
|
Bjelica B, Bartels MB, Hesebeck-Brinckmann J, Petri S. Non-motor symptoms in patients with amyotrophic lateral sclerosis: current state and future directions. J Neurol 2024; 271:3953-3977. [PMID: 38805053 PMCID: PMC11233299 DOI: 10.1007/s00415-024-12455-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Revised: 05/14/2024] [Accepted: 05/16/2024] [Indexed: 05/29/2024]
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disease characterized by the progressive degeneration of both upper and lower motor neurons. A defining histopathological feature in approximately 97% of all ALS cases is the accumulation of phosphorylated trans-activation response (TAR) DNA-binding protein 43 protein (pTDP-43) aggregates in the cytoplasm of neurons and glial cells within the central nervous system. Traditionally, it was believed that the accumulation of TDP-43 aggregates and subsequent neurodegeneration primarily occurs in motor neurons. However, contemporary evidence suggests that as the disease progresses, other systems and brain regions are also affected. Despite this, there has been a limited number of clinical studies assessing the non-motor symptoms in ALS patients. These studies often employ various outcome measures, resulting in a wide range of reported frequencies of non-motor symptoms in ALS patients. The importance of assessing the non-motor symptoms reflects in a fact that they have a significant impact on patients' quality of life, yet they frequently go underdiagnosed and unreported during clinical evaluations. This review aims to provide an up-to-date overview of the current knowledge concerning non-motor symptoms in ALS. Furthermore, we address their diagnosis and treatment in everyday clinical practice.
Collapse
Affiliation(s)
- Bogdan Bjelica
- Department of Neurology, Hannover Medical School, 1, Carl-Neuberg-Strasse, 30625, Hannover, Germany.
| | - Maj-Britt Bartels
- Precision Neurology of Neuromuscular and Motoneuron Diseases, University of Luebeck, Lübeck, Germany
| | - Jasper Hesebeck-Brinckmann
- Neurology Department, Division for Neurodegenerative Diseases, University Medicine Mannheim, Heidelberg University, Mannheim Center for Translational Medicine, Mannheim, Germany
| | - Susanne Petri
- Department of Neurology, Hannover Medical School, 1, Carl-Neuberg-Strasse, 30625, Hannover, Germany
| |
Collapse
|
10
|
Abstract
The blood-brain barrier (BBB) is a critical interface separating the central nervous system from the peripheral circulation, ensuring brain homeostasis and function. Recent research has unveiled a profound connection between the BBB and circadian rhythms, the endogenous oscillations synchronizing biological processes with the 24-hour light-dark cycle. This review explores the significance of circadian rhythms in the context of BBB functions, with an emphasis on substrate passage through the BBB. Our discussion includes efflux transporters and the molecular timing mechanisms that regulate their activities. A significant focus of this review is the potential implications of chronotherapy, leveraging our knowledge of circadian rhythms for improving drug delivery to the brain. Understanding the temporal changes in BBB can lead to optimized timing of drug administration, to enhance therapeutic efficacy for neurological disorders while reducing side effects. By elucidating the interplay between circadian rhythms and drug transport across the BBB, this review offers insights into innovative therapeutic interventions.
Collapse
Affiliation(s)
- Mari Kim
- Cell Biology Department, Emory University, Atlanta, GA, USA
| | | | | |
Collapse
|
11
|
Feigl B, Lewis SJG, Rawashdeh O. Targeting sleep and the circadian system as a novel treatment strategy for Parkinson's disease. J Neurol 2024; 271:1483-1491. [PMID: 37943299 PMCID: PMC10896880 DOI: 10.1007/s00415-023-12073-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 10/12/2023] [Accepted: 10/18/2023] [Indexed: 11/10/2023]
Abstract
There is a growing appreciation of the wide range of sleep-wake disturbances that occur frequently in Parkinson's disease. These are known to be associated with a range of motor and non-motor symptoms and significantly impact not only on the quality of life of the patient, but also on their bed partner. The underlying causes for fragmented sleep and daytime somnolence are no doubt multifactorial but there is clear evidence for circadian disruption in Parkinson's disease. This appears to be occurring not only as a result of the neuropathological changes that occur across a distributed neural network, but even down to the cellular level. Such observations indicate that circadian changes may in fact be a driver of neurodegeneration, as well as a cause for some of the sleep-wake symptoms observed in Parkinson's disease. Thus, efforts are now required to evaluate approaches including the prescription of precision medicine to modulate photoreceptor activation ratios that reflect daylight inputs to the circadian pacemaker, the use of small molecules to target clock genes, the manipulation of orexin pathways that could help restore the circadian system, to offer novel symptomatic and novel disease modifying strategies.
Collapse
Affiliation(s)
- Beatrix Feigl
- Centre for Vision and Eye Research, Queensland University of Technology (QUT), Brisbane, QLD, 4059, Australia
- School of Biomedical Sciences, Queensland University of Technology (QUT), Brisbane, QLD, 4059, Australia
- Queensland Eye Institute, South Brisbane, QLD, 4101, Australia
| | - Simon J G Lewis
- Parkinson's Disease Research Clinic, Brain and Mind Centre, School of Medical Sciences, University of Sydney, Camperdown, NSW, 2006, Australia.
| | - Oliver Rawashdeh
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, Brisbane, QLD, 4072, Australia
| |
Collapse
|
12
|
Li Y, Tan Y, Zhao Z. Impacts of aging on circadian rhythm and related sleep disorders. Biosystems 2024; 236:105111. [PMID: 38159672 DOI: 10.1016/j.biosystems.2023.105111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 12/22/2023] [Accepted: 12/22/2023] [Indexed: 01/03/2024]
Abstract
Circadian rhythm is an essential component of biology that organizes the internal synchrony of the organism in response to the environment. Aging significantly impacts circadian rhythm and is also associated with specific sleep complaints in mammals, including earlier awakening and decreased sleep consolidation at the end of the night. However, the regulation mechanism of aging on the circadian rhythm is far from clear. To further understand the impact of aging, we use an existing mathematical model of circadian rhythm combined with the aging system to explore the effects of aging on circadian rhythm and two kinds of sleep disorders, familial late sleep syndrome (FASPS) and delayed sleep syndrome (DSPS). We get a few intriguing findings from numerical simulations. Aging weakens rhythmicity by reducing the amplitude of circadian rhythm. Aging exacerbates the sleep pattern of being early to bed and early to rise by shortening the period of circadian rhythm and advancing the entrainment phase. Aging reduces the ability of the circadian rhythm to respond to light. The elderly need stronger light to get entrainment with the environmental light cycle. It is more difficult for the elderly to recover from disturbed light. Especially elderly people take a longer time to overcome jet lag. Aging worsens the "morningness" of FASPS disorder patients and improves the symptoms of DSPS disorder patients. This study helps to better understand the impacts of aging on circadian rhythm and sleep disorders and provides theoretical support for the treatment of circadian disorders in the elderly.
Collapse
Affiliation(s)
- Ying Li
- College of Information Technology, Shanghai Ocean University, Shanghai 201306, China.
| | - YuanYuan Tan
- College of Information Technology, Shanghai Ocean University, Shanghai 201306, China
| | - Zhao Zhao
- College of Information Technology, Shanghai Ocean University, Shanghai 201306, China
| |
Collapse
|
13
|
Lee DY, Jung I, Park SY, Yu JH, Seo JA, Kim KJ, Kim NH, Yoo HJ, Kim SG, Choi KM, Baik SH, Kim NH. Attention to Innate Circadian Rhythm and the Impact of Its Disruption on Diabetes. Diabetes Metab J 2024; 48:37-52. [PMID: 38173377 PMCID: PMC10850272 DOI: 10.4093/dmj.2023.0193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Accepted: 10/16/2023] [Indexed: 01/05/2024] Open
Abstract
Novel strategies are required to reduce the risk of developing diabetes and/or clinical outcomes and complications of diabetes. In this regard, the role of the circadian system may be a potential candidate for the prevention of diabetes. We reviewed evidence from animal, clinical, and epidemiological studies linking the circadian system to various aspects of the pathophysiology and clinical outcomes of diabetes. The circadian clock governs genetic, metabolic, hormonal, and behavioral signals in anticipation of cyclic 24-hour events through interactions between a "central clock" in the suprachiasmatic nucleus and "peripheral clocks" in the whole body. Currently, circadian rhythmicity in humans can be subjectively or objectively assessed by measuring melatonin and glucocorticoid levels, core body temperature, peripheral blood, oral mucosa, hair follicles, rest-activity cycles, sleep diaries, and circadian chronotypes. In this review, we summarized various circadian misalignments, such as altered light-dark, sleep-wake, rest-activity, fasting-feeding, shift work, evening chronotype, and social jetlag, as well as mutations in clock genes that could contribute to the development of diabetes and poor glycemic status in patients with diabetes. Targeting critical components of the circadian system could deliver potential candidates for the treatment and prevention of type 2 diabetes mellitus in the future.
Collapse
Affiliation(s)
- Da Young Lee
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Korea University College of Medicine, Seoul, Korea
| | - Inha Jung
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Korea University College of Medicine, Seoul, Korea
| | - So Young Park
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Korea University College of Medicine, Seoul, Korea
| | - Ji Hee Yu
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Korea University College of Medicine, Seoul, Korea
| | - Ji A Seo
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Korea University College of Medicine, Seoul, Korea
| | - Kyeong Jin Kim
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Korea University College of Medicine, Seoul, Korea
| | - Nam Hoon Kim
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Korea University College of Medicine, Seoul, Korea
| | - Hye Jin Yoo
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Korea University College of Medicine, Seoul, Korea
| | - Sin Gon Kim
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Korea University College of Medicine, Seoul, Korea
| | - Kyung Mook Choi
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Korea University College of Medicine, Seoul, Korea
| | - Sei Hyun Baik
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Korea University College of Medicine, Seoul, Korea
| | - Nan Hee Kim
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Korea University College of Medicine, Seoul, Korea
- BK21 FOUR R&E Center for Learning Health Systems, Korea University, Seoul, Korea
| |
Collapse
|
14
|
Qamar MA, Tall P, van Wamelen D, Wan YM, Rukavina K, Fieldwalker A, Matthew D, Leta V, Bannister K, Chaudhuri KR. Setting the clinical context to non-motor symptoms reflected by Park-pain, Park-sleep, and Park-autonomic subtypes of Parkinson's disease. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2023; 174:1-58. [PMID: 38341227 DOI: 10.1016/bs.irn.2023.08.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/12/2024]
Abstract
Non-motor symptoms (NMS) of Parkinson's disease (PD) are well described in both clinical practice and the literature, enabling their management and enhancing our understanding of PD. NMS can dominate the clinical pictures and NMS subtypes have recently been proposed, initially based on clinical observations, and later confirmed in data driven analyses of large datasets and in biomarker-based studies. In this chapter, we provide an update on what is known about three common subtypes of NMS in PD. The pain (Park-pain), sleep dysfunction (Park-sleep), and autonomic dysfunction (Park-autonomic), providing an overview of their individual classification, clinical manifestation, pathophysiology, diagnosis, and potential treatments.
Collapse
Affiliation(s)
- Mubasher A Qamar
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology, and Neuroscience, King's College London, London, United Kingdom; Parkinson's Foundation Centre of Excellence and Department of Neurology and Neurosciences, King's College Hospital NHS Trust, London, United Kingdom.
| | - Phoebe Tall
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology, and Neuroscience, King's College London, London, United Kingdom; Parkinson's Foundation Centre of Excellence and Department of Neurology and Neurosciences, King's College Hospital NHS Trust, London, United Kingdom
| | - Daniel van Wamelen
- Parkinson's Foundation Centre of Excellence and Department of Neurology and Neurosciences, King's College Hospital NHS Trust, London, United Kingdom; Department of Neuroimaging, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, United Kingdom; Radboud University Medical Center, Donders Institute for Brain, Cognition and Behaviour, Department of Neurology, Centre of Expertise for Parkinson & Movement Disorders, Nijmegen, The Netherlands
| | - Yi Min Wan
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology, and Neuroscience, King's College London, London, United Kingdom; Parkinson's Foundation Centre of Excellence and Department of Neurology and Neurosciences, King's College Hospital NHS Trust, London, United Kingdom; Department of Psychiatry, Ng Teng Fong General Hospital, Singapore, Singapore
| | - Katarina Rukavina
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology, and Neuroscience, King's College London, London, United Kingdom; Parkinson's Foundation Centre of Excellence and Department of Neurology and Neurosciences, King's College Hospital NHS Trust, London, United Kingdom
| | - Anna Fieldwalker
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology, and Neuroscience, King's College London, London, United Kingdom; Central Modulation of Pain Lab, Wolfson Centre for Age-Related Diseases, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom
| | - Donna Matthew
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology, and Neuroscience, King's College London, London, United Kingdom; Parkinson's Foundation Centre of Excellence and Department of Neurology and Neurosciences, King's College Hospital NHS Trust, London, United Kingdom
| | - Valentina Leta
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology, and Neuroscience, King's College London, London, United Kingdom; Parkinson's Foundation Centre of Excellence and Department of Neurology and Neurosciences, King's College Hospital NHS Trust, London, United Kingdom; Department of Clinical Neurosciences, Parkinson, and Movement Disorders Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Kirsty Bannister
- Central Modulation of Pain Lab, Wolfson Centre for Age-Related Diseases, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom
| | - K Ray Chaudhuri
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology, and Neuroscience, King's College London, London, United Kingdom; Parkinson's Foundation Centre of Excellence and Department of Neurology and Neurosciences, King's College Hospital NHS Trust, London, United Kingdom
| |
Collapse
|
15
|
Keihani A, Mayeli A, Ferrarelli F. Circadian Rhythm Changes in Healthy Aging and Mild Cognitive Impairment. Adv Biol (Weinh) 2023; 7:e2200237. [PMID: 36403250 PMCID: PMC10199146 DOI: 10.1002/adbi.202200237] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 11/01/2022] [Indexed: 11/21/2022]
Abstract
Disruptions in circadian rhythms can occur in healthy aging; however, these changes are more severe and pervasive in individuals with age-related and neurodegenerative diseases, such as dementia. Circadian rhythm alterations are also present in preclinical stages of dementia, for example, in patients with mild cognitive impairments (MCI); thus, providing a unique window of opportunity for early intervention in neurodegenerative disorders. Nonetheless, there is a lack of studies examining the association between relevant changes in circadian rhythms and their relationship with cognitive dysfunctions in MCI individuals. In this review, circadian system alterations occurring in MCI patients are examined compared to healthy aging individuals while also considering their association with MCI neurocognitive alterations. The main findings are that abnormal circadian changes in rest-activity, core body temperature, melatonin, and cortisol rhythms appear in the MCI stage and that these circadian rhythm disruptions are associated with some of the neurocognitive deficits observed in MCI patients. In addition, preliminary evidence indicates that interventions aimed at restoring regular circadian rhythms may prevent or halt the progress of neurodegenerative diseases and mitigate their related cognitive impairments. Future longitudinal studies with repeated follow-up assessments are needed to establish the translational potential of these findings in clinical practice.
Collapse
Affiliation(s)
- Ahmadreza Keihani
- Department of Psychiatry, University of Pittsburgh, 3501 Forbes Ave, Pittsburgh, PA, 15213, USA
| | - Ahmad Mayeli
- Department of Psychiatry, University of Pittsburgh, 3501 Forbes Ave, Pittsburgh, PA, 15213, USA
| | - Fabio Ferrarelli
- Department of Psychiatry, University of Pittsburgh, 3501 Forbes Ave, Pittsburgh, PA, 15213, USA
| |
Collapse
|
16
|
Gao X, Sun H, Hao S, Sun H, Ge J. Melatonin protects HT-22 cells against palmitic acid-induced glucolipid metabolic dysfunction and cell injuries: Involved in the regulation of synaptic plasticity and circadian rhythms. Biochem Pharmacol 2023; 217:115846. [PMID: 37804870 DOI: 10.1016/j.bcp.2023.115846] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 10/01/2023] [Accepted: 10/04/2023] [Indexed: 10/09/2023]
Abstract
Melatonin (MLT) is ahormonal substance reported with various pharmacological activities.Based on its effects of neuroprotection and metabolic regulation, the aim of the present study is to investigate its potential effect on palmitic acid (PA)-induced cell injuries and glucolipid metabolic dysfunction and explore the possible mechanism. Briefly, HT-22 cells were challenged with PA (0.1 mM, 24 h) and treated with MLT (10-6-10-8 mol/L). Cell proliferation, lipid accumulation and glucose consumption were detected. The protein expression of key molecular involved with the function of synaptic plasticity and circadian rhythms were measured via western blotting, and the expression of Map-2, MT1A, MT1B and Bmal1 were measured via immunofluorescence staining. The results showed that MLT could alleviate the neurotoxicity induced by PA, as indicated by the increased cell proliferation, enhanced fluorescence intensity of Map-2, and decreased lipid deposition and insulin resistance. Moreover, treatment of MLT could reverse the imbalanced expression of p-Akt, p-ERK, Synapsin I, Synaptotagmin I, BDNF, MT1B, Bmal1, and Clock in PA-induced HT-22 cells. These results suggested a remarkably neuroprotective effect of MLT against PA-induced cell injury and glucolipid metabolic dysfunction, the mechanism of which might be involved in the regulation of synaptic plasticity and circadian rhythms.
Collapse
Affiliation(s)
- Xinran Gao
- School of Pharmacy, Anhui Medical University, 81 Meishan Road, Hefei 230032, China; The Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Medical University, Hefei, China; Anhui Provincial Laboratory of Inflammatory and Immune Disease, Anhui Institute of Innovative Drugs, Hefei, China
| | - Huaizhi Sun
- School of Pharmacy, Anhui Medical University, 81 Meishan Road, Hefei 230032, China; The Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Medical University, Hefei, China; Anhui Provincial Laboratory of Inflammatory and Immune Disease, Anhui Institute of Innovative Drugs, Hefei, China
| | - Shengwei Hao
- School of Pharmacy, Anhui Medical University, 81 Meishan Road, Hefei 230032, China; The Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Medical University, Hefei, China; Anhui Provincial Laboratory of Inflammatory and Immune Disease, Anhui Institute of Innovative Drugs, Hefei, China
| | - Huimin Sun
- School of Pharmacy, Anhui Medical University, 81 Meishan Road, Hefei 230032, China; The Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Medical University, Hefei, China; Anhui Provincial Laboratory of Inflammatory and Immune Disease, Anhui Institute of Innovative Drugs, Hefei, China
| | - Jinfang Ge
- School of Pharmacy, Anhui Medical University, 81 Meishan Road, Hefei 230032, China; The Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Medical University, Hefei, China; Anhui Provincial Laboratory of Inflammatory and Immune Disease, Anhui Institute of Innovative Drugs, Hefei, China.
| |
Collapse
|
17
|
Silva EHA, Santana NNM, Seixas NRM, Bezerra LLF, Silva MMO, Santos SF, Cavalcante JS, Leocadio-Miguel MA, Engelberth RC. Blue light exposure-dependent improvement in robustness of circadian rest-activity rhythm in aged rats. PLoS One 2023; 18:e0292342. [PMID: 37792859 PMCID: PMC10550138 DOI: 10.1371/journal.pone.0292342] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Accepted: 09/18/2023] [Indexed: 10/06/2023] Open
Abstract
The aging effects on circadian rhythms have diverse implications including changes in the pattern of rhythmic expressions, such as a wide fragmentation of the rhythm of rest-activity and decrease in amplitude of activity regulated by the suprachiasmatic nucleus (SCN). The study of blue light on biological aspects has received great current interest due, among some aspects, to its positive effects on psychiatric disorders in humans. This study aims to evaluate the effect of blue light therapy on the SCN functional aspects, through the evaluation of the rest-activity rhythm, in aging rats. For this, 33 sixteen-months-old male Wistar rats underwent continuous records of locomotor activity and were exposed to periods of 6 hours of blue light during the first half of the light phase (Zeitgeber times 0-6) for 14 days. After this, the rats were maintained at 12h:12h light:dark cycle to check the long-term effect of blue light for 14 days. Blue light repeated exposure showed positive effects on the rhythmic variables of locomotor activity in aged rats, particularly the increase in amplitude, elevation of rhythmic robustness, phase advance in acrophase, and greater consolidation of the resting phase. This effect depends on the presence of daily blue light exposure. In conclusion, our results indicate that blue light is a reliable therapy to reduce circadian dysfunctions in aged rats, but other studies assessing how blue light modulates the neural components to modulate this response are still needed.
Collapse
Affiliation(s)
- Eryck Holmes A. Silva
- Laboratory of Neurochemical Studies, Department of Physiology and Behavior, Biosciences Center, Federal University of Rio Grande do Norte, Natal, Brazil
| | | | - Narita Renata M. Seixas
- Laboratory of Neurochemical Studies, Department of Physiology and Behavior, Biosciences Center, Federal University of Rio Grande do Norte, Natal, Brazil
| | - Lyzandro Lucas F. Bezerra
- Laboratory of Neurochemical Studies, Department of Physiology and Behavior, Biosciences Center, Federal University of Rio Grande do Norte, Natal, Brazil
| | - Maria Milena O. Silva
- Laboratory of Neurochemical Studies, Department of Physiology and Behavior, Biosciences Center, Federal University of Rio Grande do Norte, Natal, Brazil
| | - Sâmarah F. Santos
- Laboratory of Neurochemical Studies, Department of Physiology and Behavior, Biosciences Center, Federal University of Rio Grande do Norte, Natal, Brazil
| | - Jeferson S. Cavalcante
- Laboratory of Neurochemical Studies, Department of Physiology and Behavior, Biosciences Center, Federal University of Rio Grande do Norte, Natal, Brazil
| | | | - Rovena Clara Engelberth
- Laboratory of Neurochemical Studies, Department of Physiology and Behavior, Biosciences Center, Federal University of Rio Grande do Norte, Natal, Brazil
| |
Collapse
|
18
|
Pehlivan S. The circadian systems genes and their importance of human health. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2023; 137:1-15. [PMID: 37709372 DOI: 10.1016/bs.apcsb.2023.02.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/16/2023]
Abstract
The circadian rhythm is the timing mechanism that creates approximately 24-hour rhythms in cellular and bodily functions in almost all living species. These internal clock systems enable living organisms to predict and respond to daily changes in their environment, optimizing temporal physiology and behavior. Circadian rhythms are regulated by both genetic and environmental risk factors. Circadian rhythms play an important role in maintaining homeostasis at the systemic and tissue levels. Disruption of this rhythm lays the groundwork for human health and disease. Disruption in these rhythms increases the susceptibility to many diseases, such as cancer, psychiatric disorders, and neurodegenerative diseases. In this chapter, the characteristics of circadian rhythm and its relationship with diseases will be discussed.
Collapse
Affiliation(s)
- S Pehlivan
- Department of Medical Biology, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey.
| |
Collapse
|
19
|
Petkovic M, Yalçin M, Heese O, Relógio A. Differential expression of the circadian clock network correlates with tumour progression in gliomas. BMC Med Genomics 2023; 16:154. [PMID: 37400829 DOI: 10.1186/s12920-023-01585-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Accepted: 06/19/2023] [Indexed: 07/05/2023] Open
Abstract
BACKGROUND Gliomas are tumours arising mostly from astrocytic or oligodendrocytic precursor cells. These tumours are classified according to the updated WHO classification from 2021 in 4 grades depending on molecular and histopathological criteria. Despite novel multimodal therapeutic approaches, the vast majority of gliomas (WHO grade III and IV) are not curable. The circadian clock is an important regulator of numerous cellular processes and its dysregulation had been found during the progression of many cancers, including gliomas. RESULTS In this study, we explore expression patterns of clock-controlled genes in low-grade glioma (LGG) and glioblastoma multiforme (GBM) and show that a set of 45 clock-controlled genes can be used to distinguish GBM from normal tissue. Subsequent analysis identified 17 clock-controlled genes with a significant association with survival. The results point to a loss of correlation strength within elements of the circadian clock network in GBM compared to LGG. We further explored the progression patterns of mutations in LGG and GBM, and showed that tumour suppressor APC is lost late both in LGG and GBM. Moreover, HIF1A, involved in cellular response to hypoxia, exhibits subclonal losses in LGG, and TERT, involved in the formation of telomerase, is lost late in the GBM progression. By examining multi-sample LGG data, we find that the clock-controlled driver genes APC, HIF1A, TERT and TP53 experience frequent subclonal gains and losses. CONCLUSIONS Our results show a higher level of disrgulation at the gene expression level in GBM compared to LGG, and indicate an association between the differentially expressed clock-regulated genes and patient survival in both LGG and GBM. By reconstructing the patterns of progression in LGG and GBM, our data reveals the relatively late gains and losses of clock-regulated glioma drivers. Our analysis emphasizes the role of clock-regulated genes in glioma development and progression. Yet, further research is needed to asses their value in the development of new treatments.
Collapse
Affiliation(s)
- Marina Petkovic
- Institute for Theoretical Biology (ITB), Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, 10117, Berlin, Germany
| | - Müge Yalçin
- Institute for Theoretical Biology (ITB), Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, 10117, Berlin, Germany
- Molecular Cancer Research Center (MKFZ), Medical Department of Hematology, Oncology, and Tumour Immunology, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, 10117, Berlin, Germany
- Institute for Systems Medicine, Faculty of Human Medicine, MSH Medical School Hamburg, 20457, Hamburg, Germany
| | - Oliver Heese
- Department of Neurosurgery and Spinal Surgery, HELIOS Medical Center Schwerin, University Campus of MSH Medical School Hamburg, 20457, Hamburg, Germany
| | - Angela Relógio
- Institute for Theoretical Biology (ITB), Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, 10117, Berlin, Germany.
- Molecular Cancer Research Center (MKFZ), Medical Department of Hematology, Oncology, and Tumour Immunology, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, 10117, Berlin, Germany.
- Institute for Systems Medicine, Faculty of Human Medicine, MSH Medical School Hamburg, 20457, Hamburg, Germany.
| |
Collapse
|
20
|
Kim HJ, Kim REY, Kim S, Lee SK, Lee HW, Shin C. Earlier chronotype in midlife as a predictor of accelerated brain aging: a population-based longitudinal cohort study. Sleep 2023; 46:zsad108. [PMID: 37061816 DOI: 10.1093/sleep/zsad108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2022] [Revised: 02/28/2023] [Indexed: 04/17/2023] Open
Abstract
STUDY OBJECTIVES Evidence suggests that sleep-wake cycle disruption could be an early manifestation of neurodegeneration and might even be a risk factor for developing diseases in healthy adults. We investigated the impact of circadian phase change on structural and functional brain deterioration in a late-adulthood population. METHODS We analyzed the data of 1874 participants (mean age 58.6 ± 6.3 years, 50.3% female) from the Korean Genome and Epidemiology Study, who were identified as cognitively unimpaired. The mid-sleep time on free days corrected for sleep debt on workdays (MSFsc) at baseline was adopted as an indicator of the chronotype and used to categorize the participants into three groups. The relationships between the chronotype and longitudinal changes in the gray matter volume (GMV) and cognitive function were investigated (mean interval: 4.2 ± 0.5 years). RESULTS The mean MSFsc of the participants was 2:45 am. The earlier MSFsc was linearly associated with smaller right entorhinal GMV (β [SE] = 0.02 [0.01]; p = .001) and lower visual memory function test scores at baseline. Longitudinally, the earlier MSFsc at baseline was only significantly associated with more rapid atrophy in the temporal lobe (β [SE] = 0.18 [0.07]; p = .018) and not with other brain lobes or subregions. Moreover, the earlier MSFsc was associated with more deteriorated verbal learning and visual memory function test scores. CONCLUSIONS An earlier chronotype in midlife, measured using a questionnaire, can be a valuable indicator for individuals who should be closely monitored for the development of neurodegenerative disorders.
Collapse
Affiliation(s)
- Hyeon Jin Kim
- Department of Neurology, Korea University Ansan Hospital, Ansan, Gyeonggi, Republic of Korea
- Departments of Neurology and Medical Science, Ewha Womans University School of Medicine and Ewha Medical Research Institute, Seoul, Republic of Korea
| | - Regina E Y Kim
- Institute of Human Genomic Study, College of Medicine, Korea University, Ansan Hospital, Ansan, Gyeonggi, Republic of Korea
- Department of Psychiatry, University of Iowa, Iowa City, Iowa, USA
| | - Soriul Kim
- Institute of Human Genomic Study, College of Medicine, Korea University, Ansan Hospital, Ansan, Gyeonggi, Republic of Korea
- Division of Sleep Medicine, Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Seung Ku Lee
- Institute of Human Genomic Study, College of Medicine, Korea University, Ansan Hospital, Ansan, Gyeonggi, Republic of Korea
| | - Hyang Woon Lee
- Departments of Neurology and Medical Science, Ewha Womans University School of Medicine and Ewha Medical Research Institute, Seoul, Republic of Korea
- Computational Medicine, Graduate Programs in System Health Science & Engineering and Artificial Intelligence Convergence, Ewha Womans University, Seoul, Republic of Korea
| | - Chol Shin
- Institute of Human Genomic Study, College of Medicine, Korea University, Ansan Hospital, Ansan, Gyeonggi, Republic of Korea
- Biomedical Research Center, Korea University Ansan Hospital, Ansan, Gyeonggi, Republic of Korea
- College of Medicine, Korea University, Seoul, Republic of Korea
| |
Collapse
|
21
|
Santos CRA, Cipolla-Neto J, Krohn M, Gonçalves I, Quintela T. Editorial: Crossing brain barriers in health and disease: Impact of circadian rhythms. Front Neurosci 2023; 17:1176084. [PMID: 37051143 PMCID: PMC10083483 DOI: 10.3389/fnins.2023.1176084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Accepted: 03/14/2023] [Indexed: 03/29/2023] Open
Affiliation(s)
- Cecília R. A. Santos
- CICS-UBI - Health Sciences Research Centre, University of Beira Interior, Covilhã, Portugal
| | - José Cipolla-Neto
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | | | - Isabel Gonçalves
- CICS-UBI - Health Sciences Research Centre, University of Beira Interior, Covilhã, Portugal
| | - Telma Quintela
- CICS-UBI - Health Sciences Research Centre, University of Beira Interior, Covilhã, Portugal
- UDI-IPG - Unidade de Investigação para o Desenvolvimento do Interior, Instituto Politécnico da Guarda, Guarda, Portugal
- *Correspondence: Telma Quintela
| |
Collapse
|
22
|
Richardson RB, Mailloux RJ. Mitochondria Need Their Sleep: Redox, Bioenergetics, and Temperature Regulation of Circadian Rhythms and the Role of Cysteine-Mediated Redox Signaling, Uncoupling Proteins, and Substrate Cycles. Antioxidants (Basel) 2023; 12:antiox12030674. [PMID: 36978924 PMCID: PMC10045244 DOI: 10.3390/antiox12030674] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2023] [Revised: 02/27/2023] [Accepted: 02/28/2023] [Indexed: 03/12/2023] Open
Abstract
Although circadian biorhythms of mitochondria and cells are highly conserved and crucial for the well-being of complex animals, there is a paucity of studies on the reciprocal interactions between oxidative stress, redox modifications, metabolism, thermoregulation, and other major oscillatory physiological processes. To address this limitation, we hypothesize that circadian/ultradian interaction of the redoxome, bioenergetics, and temperature signaling strongly determine the differential activities of the sleep–wake cycling of mammalians and birds. Posttranslational modifications of proteins by reversible cysteine oxoforms, S-glutathionylation and S-nitrosylation are shown to play a major role in regulating mitochondrial reactive oxygen species production, protein activity, respiration, and metabolomics. Nuclear DNA repair and cellular protein synthesis are maximized during the wake phase, whereas the redoxome is restored and mitochondrial remodeling is maximized during sleep. Hence, our analysis reveals that wakefulness is more protective and restorative to the nucleus (nucleorestorative), whereas sleep is more protective and restorative to mitochondria (mitorestorative). The “redox–bioenergetics–temperature and differential mitochondrial–nuclear regulatory hypothesis” adds to the understanding of mitochondrial respiratory uncoupling, substrate cycling control and hibernation. Similarly, this hypothesis explains how the oscillatory redox–bioenergetics–temperature–regulated sleep–wake states, when perturbed by mitochondrial interactome disturbances, influence the pathogenesis of aging, cancer, spaceflight health effects, sudden infant death syndrome, and diseases of the metabolism and nervous system.
Collapse
Affiliation(s)
- Richard B. Richardson
- Radiobiology and Health, Canadian Nuclear Laboratories (CNL), Chalk River, ON K0J 1J0, Canada
- McGill Medical Physics Unit, Cedars Cancer Centre—Glen Site, McGill University, Montreal, QC H4A 3J1, Canada
- Correspondence: or
| | - Ryan J. Mailloux
- School of Human Nutrition, Faculty of Agricultural and Environmental Sciences, McGill University, Sainte-Anne-de-Bellevue, QC H9X 3V9, Canada;
| |
Collapse
|
23
|
Schurhoff N, Toborek M. Circadian rhythms in the blood-brain barrier: impact on neurological disorders and stress responses. Mol Brain 2023; 16:5. [PMID: 36635730 PMCID: PMC9835375 DOI: 10.1186/s13041-023-00997-0] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Accepted: 01/03/2023] [Indexed: 01/14/2023] Open
Abstract
Circadian disruption has become more prevalent in society due to the increase in shift work, sleep disruption, blue light exposure, and travel via different time zones. The circadian rhythm is a timed transcription-translation feedback loop with positive regulators, BMAL1 and CLOCK, that interact with negative regulators, CRY and PER, to regulate both the central and peripheral clocks. This review highlights the functions of the circadian rhythm, specifically in the blood-brain barrier (BBB), during both healthy and pathological states. The BBB is a highly selective dynamic interface composed of CNS endothelial cells, astrocytes, pericytes, neurons, and microglia that form the neurovascular unit (NVU). Circadian rhythms modulate BBB integrity through regulating oscillations of tight junction proteins, assisting in functions of the NVU, and modulating transporter functions. Circadian disruptions within the BBB have been observed in stress responses and several neurological disorders, including brain metastasis, epilepsy, Alzheimer's disease, and Parkinson's disease. Further understanding of these interactions may facilitate the development of improved treatment options and preventative measures.
Collapse
Affiliation(s)
- Nicolette Schurhoff
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Suite 528, 1011 NW 15th Street, Miami, FL, 33155, USA
| | - Michal Toborek
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Suite 528, 1011 NW 15th Street, Miami, FL, 33155, USA.
- Institute of Physiotherapy and Health Sciences, The Jerzy Kukuczka Academy of Physical Education, 40-065, Katowice, Poland.
| |
Collapse
|
24
|
Shukla M, Vincent B. Melatonin as a Harmonizing Factor of Circadian Rhythms, Neuronal Cell Cycle and Neurogenesis: Additional Arguments for Its Therapeutic Use in Alzheimer's Disease. Curr Neuropharmacol 2023; 21:1273-1298. [PMID: 36918783 PMCID: PMC10286584 DOI: 10.2174/1570159x21666230314142505] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 12/07/2022] [Accepted: 12/31/2022] [Indexed: 03/16/2023] Open
Abstract
The synthesis and release of melatonin in the brain harmonize various physiological functions. The apparent decline in melatonin levels with advanced aging is an aperture to the neurodegenerative processes. It has been indicated that down regulation of melatonin leads to alterations of circadian rhythm components, which further causes a desynchronization of several genes and results in an increased susceptibility to develop neurodegenerative diseases. Additionally, as circadian rhythms and memory are intertwined, such rhythmic disturbances influence memory formation and recall. Besides, cell cycle events exhibit a remarkable oscillatory system, which is downstream of the circadian phenomena. The linkage between the molecular machinery of the cell cycle and complex fundamental regulatory proteins emphasizes the conjectural regulatory role of cell cycle components in neurodegenerative disorders such as Alzheimer's disease. Among the mechanisms intervening long before the signs of the disease appear, the disturbances of the circadian cycle, as well as the alteration of the machinery of the cell cycle and impaired neurogenesis, must hold our interest. Therefore, in the present review, we propose to discuss the underlying mechanisms of action of melatonin in regulating the circadian rhythm, cell cycle components and adult neurogenesis in the context of AD pathogenesis with the view that it might further assist to identify new therapeutic targets.
Collapse
Affiliation(s)
- Mayuri Shukla
- Institute of Molecular Biosciences, Mahidol University, Nakhon Pathom 73170, Thailand
- Present Address: Chulabhorn Graduate Institute, Chulabhorn Royal Academy, 10210, Bangkok, Thailand
| | - Bruno Vincent
- Institute of Molecular Biosciences, Mahidol University, Nakhon Pathom 73170, Thailand
- Institute of Molecular and Cellular Pharmacology, Laboratory of Excellence DistALZ, Université Côte d'Azur, INSERM, CNRS, Sophia-Antipolis, 06560, Valbonne, France
| |
Collapse
|
25
|
Richardson RB, Mailloux RJ. WITHDRAWN: Mitochondria need their sleep: Sleep-wake cycling and the role of redox, bioenergetics, and temperature regulation, involving cysteine-mediated redox signaling, uncoupling proteins, and substrate cycles. Free Radic Biol Med 2022:S0891-5849(22)01013-9. [PMID: 36462628 DOI: 10.1016/j.freeradbiomed.2022.11.036] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Accepted: 11/25/2022] [Indexed: 12/03/2022]
Abstract
This article has been withdrawn at the request of the author(s) and/or editor. The Publisher apologizes for any inconvenience this may cause. The full Elsevier Policy on Article Withdrawal can be found at https://www.elsevier.com/about/our-business/policies/article-withdrawal
Collapse
Affiliation(s)
- Richard B Richardson
- Radiobiology and Health, Canadian Nuclear Laboratories (CNL), Chalk River Laboratories, Chalk River, Ontario, K0J 1J0, Canada; McGill Medical Physics Unit, McGill University, Cedars Cancer Centre - Glen Site, Montreal, Quebec QC, H4A 3J1, Canada.
| | - Ryan J Mailloux
- School of Human Nutrition, Faculty of Agricultural and Environmental Sciences, McGill University, Sainte-Anne-de-Bellevue, Quebec, H9X 3V9, Canada
| |
Collapse
|
26
|
Antikainen E, Njoum H, Kudelka J, Branco D, Rehman RZU, Macrae V, Davies K, Hildesheim H, Emmert K, Reilmann R, Janneke van der Woude C, Maetzler W, Ng WF, O’Donnell P, Van Gassen G, Baribaud F, Pandis I, Manyakov NV, van Gils M, Ahmaniemi T, Chatterjee M. Assessing fatigue and sleep in chronic diseases using physiological signals from wearables: A pilot study. Front Physiol 2022; 13:968185. [PMID: 36452041 PMCID: PMC9702812 DOI: 10.3389/fphys.2022.968185] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Accepted: 10/31/2022] [Indexed: 08/07/2023] Open
Abstract
Problems with fatigue and sleep are highly prevalent in patients with chronic diseases and often rated among the most disabling symptoms, impairing their activities of daily living and the health-related quality of life (HRQoL). Currently, they are evaluated primarily via Patient Reported Outcomes (PROs), which can suffer from recall biases and have limited sensitivity to temporal variations. Objective measurements from wearable sensors allow to reliably quantify disease state, changes in the HRQoL, and evaluate therapeutic outcomes. This work investigates the feasibility of capturing continuous physiological signals from an electrocardiography-based wearable device for remote monitoring of fatigue and sleep and quantifies the relationship of objective digital measures to self-reported fatigue and sleep disturbances. 136 individuals were followed for a total of 1,297 recording days in a longitudinal multi-site study conducted in free-living settings and registered with the German Clinical Trial Registry (DRKS00021693). Participants comprised healthy individuals (N = 39) and patients with neurodegenerative disorders (NDD, N = 31) and immune mediated inflammatory diseases (IMID, N = 66). Objective physiological measures correlated with fatigue and sleep PROs, while demonstrating reasonable signal quality. Furthermore, analysis of heart rate recovery estimated during activities of daily living showed significant differences between healthy and patient groups. This work underscores the promise and sensitivity of novel digital measures from multimodal sensor time-series to differentiate chronic patients from healthy individuals and monitor their HRQoL. The presented work provides clinicians with realistic insights of continuous at home patient monitoring and its practical value in quantitative assessment of fatigue and sleep, an area of unmet need.
Collapse
Affiliation(s)
- Emmi Antikainen
- VTT Technical Research Centre of Finland Ltd., Tampere, Finland
| | | | - Jennifer Kudelka
- Department of Neurology, University Hospital Schleswig-Holstein, Kiel University, Kiel, Germany
| | - Diogo Branco
- LASIGE, Faculdade de Ciências, Universidade de Lisboa, Lisbon, Portugal
| | - Rana Zia Ur Rehman
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle Upon Tyne, United Kingdom
| | - Victoria Macrae
- NIHR Newcastle Biomedical Research Centre and NIHR Newcastle Clinical Research Facility, Newcastle Upon Tyne Hospitals NHS Foundation Trust, Newcastle Upon Tyne, United Kingdom
| | - Kristen Davies
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle Upon Tyne, United Kingdom
| | - Hanna Hildesheim
- Department of Neurology, University Hospital Schleswig-Holstein, Kiel University, Kiel, Germany
| | - Kirsten Emmert
- Department of Neurology, University Hospital Schleswig-Holstein, Kiel University, Kiel, Germany
| | - Ralf Reilmann
- George-Huntington-Institute, University of Münster, Münster, Germany
- Department of Clinical Radiology, University of Münster, Münster, Germany
- Department of Neurodegenerative Diseases and Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany
| | | | - Walter Maetzler
- Department of Neurology, University Hospital Schleswig-Holstein, Kiel University, Kiel, Germany
| | - Wan-Fai Ng
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle Upon Tyne, United Kingdom
- NIHR Newcastle Biomedical Research Centre and NIHR Newcastle Clinical Research Facility, Newcastle Upon Tyne Hospitals NHS Foundation Trust, Newcastle Upon Tyne, United Kingdom
| | - Patricio O’Donnell
- Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont, MA, United States
| | | | | | | | | | - Mark van Gils
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Teemu Ahmaniemi
- VTT Technical Research Centre of Finland Ltd., Tampere, Finland
| | | |
Collapse
|
27
|
SIRT1 activation and its circadian clock control: a promising approach against (frailty in) neurodegenerative disorders. Aging Clin Exp Res 2022; 34:2963-2976. [DOI: 10.1007/s40520-022-02257-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 09/14/2022] [Indexed: 11/01/2022]
|
28
|
Mauri S, Favaro M, Bernardo G, Mazzotta GM, Ziviani E. Mitochondrial autophagy in the sleeping brain. Front Cell Dev Biol 2022; 10:956394. [PMID: 36092697 PMCID: PMC9449320 DOI: 10.3389/fcell.2022.956394] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Accepted: 07/13/2022] [Indexed: 11/13/2022] Open
Abstract
A significant percentage of the mitochondrial mass is replaced on a daily basis via mechanisms of mitochondrial quality control. Through mitophagy (a selective type of autophagy that promotes mitochondrial proteostasis) cells keep a healthy pool of mitochondria, and prevent oxidative stress and inflammation. Furthermore, mitophagy helps adapting to the metabolic demand of the cells, which changes on a daily basis.Core components of the mitophagy process are PINK1 and Parkin, which mutations are linked to Parkinson’s Disease. The crucial role of PINK1/Parkin pathway during stress-induced mitophagy has been extensively studied in vitro in different cell types. However, recent advances in the field allowed discovering that mitophagy seems to be only slightly affected in PINK1 KO mice and flies, putting into question the physiological relevance of this pathway in vivo in the whole organism. Indeed, several cell-specific PINK1/Parkin-independent mitophagy pathways have been recently discovered, which appear to be activated under physiological conditions such as those that promote mitochondrial proteome remodeling during differentiation or in response to specific physiological stimuli.In this Mini Review we want to summarize the recent advances in the field, and add another level of complexity by focusing attention on a potentially important aspect of mitophagy regulation: the implication of the circadian clock. Recent works showed that the circadian clock controls many aspects of mitochondrial physiology, including mitochondrial morphology and dynamic, respiratory activity, and ATP synthesis. Furthermore, one of the essential functions of sleep, which is controlled by the clock, is the clearance of toxic metabolic compounds from the brain, including ROS, via mechanisms of proteostasis. Very little is known about a potential role of the clock in the quality control mechanisms that maintain the mitochondrial repertoire healthy during sleep/wake cycles. More importantly, it remains completely unexplored whether (dys)function of mitochondrial proteostasis feedbacks to the circadian clockwork.
Collapse
Affiliation(s)
| | | | | | | | - Elena Ziviani
- *Correspondence: Gabriella M. Mazzotta, Elena Ziviani,
| |
Collapse
|
29
|
Salemi M, Mogavero MP, Lanza G, Mongioì LM, Calogero AE, Ferri R. Examples of Inverse Comorbidity between Cancer and Neurodegenerative Diseases: A Possible Role for Noncoding RNA. Cells 2022; 11:1930. [PMID: 35741059 PMCID: PMC9221903 DOI: 10.3390/cells11121930] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Revised: 05/25/2022] [Accepted: 06/13/2022] [Indexed: 02/06/2023] Open
Abstract
Cancer is one of the most common causes of death; in parallel, the incidence and prevalence of central nervous system diseases are equally high. Among neurodegenerative diseases, Alzheimer's dementia is the most common, while Parkinson's disease (PD) is the second most frequent neurodegenerative disease. There is a significant amount of evidence on the complex biological connection between cancer and neurodegeneration. Noncoding RNAs (ncRNAs) are defined as transcribed nucleotides that perform a variety of regulatory functions. The mechanisms by which ncRNAs exert their functions are numerous and involve every aspect of cellular life. The same ncRNA can act in multiple ways, leading to different outcomes; in fact, a single ncRNA can participate in the pathogenesis of more than one disease-even if these seem very different, as cancer and neurodegenerative disorders are. The ncRNA activates specific pathways leading to one or the other clinical phenotype, sometimes with obvious mechanisms of inverse comorbidity. We aimed to collect from the existing literature examples of inverse comorbidity in which ncRNAs seem to play a key role. We also investigated the example of mir-519a-3p, and one of its target genes Poly (ADP-ribose) polymerase 1, for the inverse comorbidity mechanism between some cancers and PD. We believe it is very important to study the inverse comorbidity relationship between cancer and neurodegenerative diseases because it will help us to better assess these two major areas of human disease.
Collapse
Affiliation(s)
- Michele Salemi
- Oasi Research Institute, IRCCS (Istituti di Ricovero e Cura a Carattere Scientifico), Italian Ministry of Health, 94018 Troina, Italy; (G.L.); (R.F.)
| | - Maria Paola Mogavero
- Istituti Clinici Scientifici Maugeri, IRCCS (Istituti di Ricovero e Cura a Carattere Scientifico), Scientific Institute of Pavia, 27100 Pavia, Italy;
| | - Giuseppe Lanza
- Oasi Research Institute, IRCCS (Istituti di Ricovero e Cura a Carattere Scientifico), Italian Ministry of Health, 94018 Troina, Italy; (G.L.); (R.F.)
- Department of Surgery and Medical-Surgical Specialties, University of Catania, 95123 Catania, Italy
| | - Laura M. Mongioì
- Department of Clinical and Experimental Medicine, University of Catania, 95123 Catania, Italy; (L.M.M.); (A.E.C.)
| | - Aldo E. Calogero
- Department of Clinical and Experimental Medicine, University of Catania, 95123 Catania, Italy; (L.M.M.); (A.E.C.)
| | - Raffaele Ferri
- Oasi Research Institute, IRCCS (Istituti di Ricovero e Cura a Carattere Scientifico), Italian Ministry of Health, 94018 Troina, Italy; (G.L.); (R.F.)
| |
Collapse
|
30
|
Prakash P, Pradhan AK, Sheeba V. Hsp40 overexpression in pacemaker neurons delays circadian dysfunction in a Drosophila model of Huntington's disease. Dis Model Mech 2022; 15:275556. [PMID: 35645202 PMCID: PMC9254228 DOI: 10.1242/dmm.049447] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Accepted: 05/24/2022] [Indexed: 12/13/2022] Open
Abstract
Circadian disturbances are early features of neurodegenerative diseases, including Huntington's disease (HD). Emerging evidence suggests that circadian decline feeds into neurodegenerative symptoms, exacerbating them. Therefore, we asked whether known neurotoxic modifiers can suppress circadian dysfunction. We performed a screen of neurotoxicity-modifier genes to suppress circadian behavioural arrhythmicity in a Drosophila circadian HD model. The molecular chaperones Hsp40 and HSP70 emerged as significant suppressors in the circadian context, with Hsp40 being the more potent mitigator. Upon Hsp40 overexpression in the Drosophila circadian ventrolateral neurons (LNv), the behavioural rescue was associated with neuronal rescue of loss of circadian proteins from small LNv soma. Specifically, there was a restoration of the molecular clock protein Period and its oscillations in young flies and a long-lasting rescue of the output neuropeptide Pigment dispersing factor. Significantly, there was a reduction in the expanded Huntingtin inclusion load, concomitant with the appearance of a spot-like Huntingtin form. Thus, we provide evidence implicating the neuroprotective chaperone Hsp40 in circadian rehabilitation. The involvement of molecular chaperones in circadian maintenance has broader therapeutic implications for neurodegenerative diseases. This article has an associated First Person interview with the first author of the paper. Summary: This study shows, for the first time, a neuroprotective role of chaperone Hsp40 in suppressing circadian dysfunction associated with Huntington's disease in a Drosophila model.
Collapse
Affiliation(s)
- Pavitra Prakash
- Evolutionary and Integrative Biology Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Bangalore 560064, India
| | - Arpit Kumar Pradhan
- Neuroscience Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Bangalore 560064, India
| | - Vasu Sheeba
- Evolutionary and Integrative Biology Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Bangalore 560064, India.,Neuroscience Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Bangalore 560064, India
| |
Collapse
|
31
|
Yalçin M, Mundorf A, Thiel F, Amatriain-Fernández S, Kalthoff IS, Beucke JC, Budde H, Garthus-Niegel S, Peterburs J, Relógio A. It's About Time: The Circadian Network as Time-Keeper for Cognitive Functioning, Locomotor Activity and Mental Health. Front Physiol 2022; 13:873237. [PMID: 35547585 PMCID: PMC9081535 DOI: 10.3389/fphys.2022.873237] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Accepted: 03/08/2022] [Indexed: 12/24/2022] Open
Abstract
A variety of organisms including mammals have evolved a 24h, self-sustained timekeeping machinery known as the circadian clock (biological clock), which enables to anticipate, respond, and adapt to environmental influences such as the daily light and dark cycles. Proper functioning of the clock plays a pivotal role in the temporal regulation of a wide range of cellular, physiological, and behavioural processes. The disruption of circadian rhythms was found to be associated with the onset and progression of several pathologies including sleep and mental disorders, cancer, and neurodegeneration. Thus, the role of the circadian clock in health and disease, and its clinical applications, have gained increasing attention, but the exact mechanisms underlying temporal regulation require further work and the integration of evidence from different research fields. In this review, we address the current knowledge regarding the functioning of molecular circuits as generators of circadian rhythms and the essential role of circadian synchrony in a healthy organism. In particular, we discuss the role of circadian regulation in the context of behaviour and cognitive functioning, delineating how the loss of this tight interplay is linked to pathological development with a focus on mental disorders and neurodegeneration. We further describe emerging new aspects on the link between the circadian clock and physical exercise-induced cognitive functioning, and its current usage as circadian activator with a positive impact in delaying the progression of certain pathologies including neurodegeneration and brain-related disorders. Finally, we discuss recent epidemiological evidence pointing to an important role of the circadian clock in mental health.
Collapse
Affiliation(s)
- Müge Yalçin
- Institute for Theoretical Biology (ITB), Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Berlin, Germany
- Molecular Cancer Research Center (MKFZ), Medical Department of Hematology, Oncology, and Tumour Immunology, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Berlin, Germany
| | - Annakarina Mundorf
- Institute for Systems Medicine and Faculty of Human Medicine, MSH Medical School Hamburg, Hamburg, Germany
| | - Freya Thiel
- Institute for Systems Medicine and Faculty of Human Medicine, MSH Medical School Hamburg, Hamburg, Germany
- Institute and Policlinic of Occupational and Social Medicine, Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
| | - Sandra Amatriain-Fernández
- Institute for Systems Medicine and Faculty of Human Sciences, MSH Medical School Hamburg, Hamburg, Germany
| | - Ida Schulze Kalthoff
- Institute for Systems Medicine and Faculty of Human Medicine, MSH Medical School Hamburg, Hamburg, Germany
| | - Jan-Carl Beucke
- Institute for Systems Medicine and Faculty of Human Medicine, MSH Medical School Hamburg, Hamburg, Germany
- Department of Psychology, Humboldt-Universität zu Berlin, Berlin, Germany
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Henning Budde
- Institute for Systems Medicine and Faculty of Human Sciences, MSH Medical School Hamburg, Hamburg, Germany
| | - Susan Garthus-Niegel
- Institute for Systems Medicine and Faculty of Human Medicine, MSH Medical School Hamburg, Hamburg, Germany
- Institute and Policlinic of Occupational and Social Medicine, Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
- Department of Child Health and Development, Norwegian Institute of Public Health, Oslo, Norway
| | - Jutta Peterburs
- Institute for Systems Medicine and Faculty of Human Medicine, MSH Medical School Hamburg, Hamburg, Germany
| | - Angela Relógio
- Institute for Theoretical Biology (ITB), Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Berlin, Germany
- Molecular Cancer Research Center (MKFZ), Medical Department of Hematology, Oncology, and Tumour Immunology, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Berlin, Germany
- Institute for Systems Medicine and Faculty of Human Medicine, MSH Medical School Hamburg, Hamburg, Germany
| |
Collapse
|
32
|
Yang C, Hwang C, Tsai N, Yang M. Expression of circadian clock genes in leukocytes of patients with Meniere's disease. Laryngoscope Investig Otolaryngol 2022; 7:584-591. [PMID: 35434324 PMCID: PMC9008173 DOI: 10.1002/lio2.757] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2021] [Revised: 01/07/2022] [Accepted: 02/01/2022] [Indexed: 11/18/2022] Open
Abstract
Objectives The underlying etiology of Meniere's disease (MD) is not completely clear, but the precipitated triggers may alter the circadian clock in patients with MD. This study aims to survey the expression of circadian clock genes in peripheral blood (PB) leukocytes of MD patients. Methods We investigated the expression of nine circadian clock genes in the PB leukocytes of patients with MD and normal controls using real‐time quantitative reverse transcriptase‐polymerase chain reaction (qRT‐PCR). Results We observed significantly lower expression of PER1 gene and higher expression of CLOCK gene in MD patients than those in normal controls (p < 0.05). PER1 did not associate with the degree of dizziness handicap in the patients with MD, but a lower expression of PER1 was significantly correlated with higher pure tone average (PTA) and speech reception threshold of the affected ear (p < 0.05). Patients with PTA > 30 dB had significantly lower PER1 expression than those with PTA ≤30 dB in the affected ear (p < 0.05). Our qRT‐PCR result was validated by fewer positively stained leukocytes for PER1 protein in the MD patients using the immunocytochemical study. Conclusion Our study implies the alteration of the circadian clock in patients with MD. In particular, the downregulation of PER1 correlated with the degree of hearing loss in the affected ear. PER1 in PB leukocytes may be a potential marker for the progression of hearing loss in MD.
Collapse
Affiliation(s)
- Chao‐Hui Yang
- Department of Otolaryngology Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine Kaohsiung Taiwan
| | - Chung‐Feng Hwang
- Department of Otolaryngology Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine Kaohsiung Taiwan
| | - Nai‐Wen Tsai
- Department of Neurology Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine Kaohsiung Taiwan
| | - Ming‐Yu Yang
- Department of Otolaryngology Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine Kaohsiung Taiwan
- Graduate Institute of Clinical Medical Sciences College of Medicine, Chang Gung University Tao‐Yuan Taiwan
| |
Collapse
|
33
|
Gabryelska A, Turkiewicz S, Karuga FF, Sochal M, Strzelecki D, Białasiewicz P. Disruption of Circadian Rhythm Genes in Obstructive Sleep Apnea Patients-Possible Mechanisms Involved and Clinical Implication. Int J Mol Sci 2022; 23:ijms23020709. [PMID: 35054894 PMCID: PMC8775490 DOI: 10.3390/ijms23020709] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 12/29/2021] [Accepted: 01/05/2022] [Indexed: 02/06/2023] Open
Abstract
Obstructive sleep apnea (OSA) is a chronic condition characterized by recurrent pauses in breathing caused by the collapse of the upper airways, which results in intermittent hypoxia and arousals during the night. The disorder is associated with a vast number of comorbidities affecting different systems, including cardiovascular, metabolic, psychiatric, and neurological complications. Due to abnormal sleep architecture, OSA patients are at high risk of circadian clock disruption, as has been reported in several recent studies. The circadian clock affects almost all daily behavioral patterns, as well as a plethora of physiological processes, and might be one of the key factors contributing to OSA complications. An intricate interaction between the circadian clock and hypoxia may further affect these processes, which has a strong foundation on the molecular level. Recent studies revealed an interaction between hypoxia-inducible factor 1 (HIF-1), a key regulator of oxygen metabolism, and elements of circadian clocks. This relationship has a strong base in the structure of involved elements, as HIF-1 as well as PER, CLOCK, and BMAL, belong to the same Per-Arnt-Sim domain family. Therefore, this review summarizes the available knowledge on the molecular mechanism of circadian clock disruption and its influence on the development and progression of OSA comorbidities.
Collapse
Affiliation(s)
- Agata Gabryelska
- Department of Sleep Medicine and Metabolic Disorders, Medical University of Lodz, 92-215 Lodz, Poland; (S.T.); (F.F.K.); (M.S.); (P.B.)
- Correspondence: ; Tel.: +48-660796004
| | - Szymon Turkiewicz
- Department of Sleep Medicine and Metabolic Disorders, Medical University of Lodz, 92-215 Lodz, Poland; (S.T.); (F.F.K.); (M.S.); (P.B.)
| | - Filip Franciszek Karuga
- Department of Sleep Medicine and Metabolic Disorders, Medical University of Lodz, 92-215 Lodz, Poland; (S.T.); (F.F.K.); (M.S.); (P.B.)
| | - Marcin Sochal
- Department of Sleep Medicine and Metabolic Disorders, Medical University of Lodz, 92-215 Lodz, Poland; (S.T.); (F.F.K.); (M.S.); (P.B.)
| | - Dominik Strzelecki
- Department of Affective and Psychotic Disorders, Medical University of Lodz, 92-215 Lodz, Poland;
| | - Piotr Białasiewicz
- Department of Sleep Medicine and Metabolic Disorders, Medical University of Lodz, 92-215 Lodz, Poland; (S.T.); (F.F.K.); (M.S.); (P.B.)
| |
Collapse
|
34
|
Hunt J, Coulson EJ, Rajnarayanan R, Oster H, Videnovic A, Rawashdeh O. Sleep and circadian rhythms in Parkinson's disease and preclinical models. Mol Neurodegener 2022; 17:2. [PMID: 35000606 PMCID: PMC8744293 DOI: 10.1186/s13024-021-00504-w] [Citation(s) in RCA: 50] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Accepted: 11/30/2021] [Indexed: 12/21/2022] Open
Abstract
The use of animals as models of human physiology is, and has been for many years, an indispensable tool for understanding the mechanisms of human disease. In Parkinson's disease, various mouse models form the cornerstone of these investigations. Early models were developed to reflect the traditional histological features and motor symptoms of Parkinson's disease. However, it is important that models accurately encompass important facets of the disease to allow for comprehensive mechanistic understanding and translational significance. Circadian rhythm and sleep issues are tightly correlated to Parkinson's disease, and often arise prior to the presentation of typical motor deficits. It is essential that models used to understand Parkinson's disease reflect these dysfunctions in circadian rhythms and sleep, both to facilitate investigations into mechanistic interplay between sleep and disease, and to assist in the development of circadian rhythm-facing therapeutic treatments. This review describes the extent to which various genetically- and neurotoxically-induced murine models of Parkinson's reflect the sleep and circadian abnormalities of Parkinson's disease observed in the clinic.
Collapse
Affiliation(s)
- Jeremy Hunt
- School of Biomedical Sciences, Faculty of Medicine, University of Queensland, Brisbane, Australia
| | - Elizabeth J. Coulson
- School of Biomedical Sciences, Faculty of Medicine, University of Queensland, Brisbane, Australia
- Queensland Brain Institute, University of Queensland, Brisbane, Australia
| | | | - Henrik Oster
- Institute of Neurobiology, University of Lübeck, Lübeck, Germany
| | - Aleksandar Videnovic
- Movement Disorders Unit and Division of Sleep Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA USA
| | - Oliver Rawashdeh
- School of Biomedical Sciences, Faculty of Medicine, University of Queensland, Brisbane, Australia
| |
Collapse
|
35
|
Peng X, Fan R, Xie L, Shi X, Dong K, Zhang S, Tao J, Xu W, Ma D, Chen J, Yang Y. A Growing Link between Circadian Rhythms, Type 2 Diabetes Mellitus and Alzheimer's Disease. Int J Mol Sci 2022; 23:504. [PMID: 35008933 PMCID: PMC8745289 DOI: 10.3390/ijms23010504] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2021] [Revised: 12/29/2021] [Accepted: 12/31/2021] [Indexed: 02/04/2023] Open
Abstract
Type 2 diabetes mellitus (T2DM) patients are at a higher risk of developing Alzheimer's disease (AD). Mounting evidence suggests the emerging important role of circadian rhythms in many diseases. Circadian rhythm disruption is considered to contribute to both T2DM and AD. Here, we review the relationship among circadian rhythm disruption, T2DM and AD, and suggest that the occurrence and progression of T2DM and AD may in part be associated with circadian disruption. Then, we summarize the promising therapeutic strategies targeting circadian dysfunction for T2DM and AD, including pharmacological treatment such as melatonin, orexin, and circadian molecules, as well as non-pharmacological treatments like light therapy, feeding behavior, and exercise.
Collapse
Affiliation(s)
- Xuemin Peng
- Department of Endocrinology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (X.P.); (R.F.); (L.X.); (X.S.); (K.D.); (S.Z.); (J.T.); (W.X.); (D.M.)
- Branch of National Clinical Research Center for Metabolic Diseases, Wuhan 430030, China
| | - Rongping Fan
- Department of Endocrinology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (X.P.); (R.F.); (L.X.); (X.S.); (K.D.); (S.Z.); (J.T.); (W.X.); (D.M.)
- Branch of National Clinical Research Center for Metabolic Diseases, Wuhan 430030, China
| | - Lei Xie
- Department of Endocrinology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (X.P.); (R.F.); (L.X.); (X.S.); (K.D.); (S.Z.); (J.T.); (W.X.); (D.M.)
- Branch of National Clinical Research Center for Metabolic Diseases, Wuhan 430030, China
| | - Xiaoli Shi
- Department of Endocrinology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (X.P.); (R.F.); (L.X.); (X.S.); (K.D.); (S.Z.); (J.T.); (W.X.); (D.M.)
- Branch of National Clinical Research Center for Metabolic Diseases, Wuhan 430030, China
| | - Kun Dong
- Department of Endocrinology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (X.P.); (R.F.); (L.X.); (X.S.); (K.D.); (S.Z.); (J.T.); (W.X.); (D.M.)
- Branch of National Clinical Research Center for Metabolic Diseases, Wuhan 430030, China
| | - Shujun Zhang
- Department of Endocrinology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (X.P.); (R.F.); (L.X.); (X.S.); (K.D.); (S.Z.); (J.T.); (W.X.); (D.M.)
- Branch of National Clinical Research Center for Metabolic Diseases, Wuhan 430030, China
| | - Jing Tao
- Department of Endocrinology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (X.P.); (R.F.); (L.X.); (X.S.); (K.D.); (S.Z.); (J.T.); (W.X.); (D.M.)
- Branch of National Clinical Research Center for Metabolic Diseases, Wuhan 430030, China
| | - Weijie Xu
- Department of Endocrinology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (X.P.); (R.F.); (L.X.); (X.S.); (K.D.); (S.Z.); (J.T.); (W.X.); (D.M.)
- Branch of National Clinical Research Center for Metabolic Diseases, Wuhan 430030, China
| | - Delin Ma
- Department of Endocrinology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (X.P.); (R.F.); (L.X.); (X.S.); (K.D.); (S.Z.); (J.T.); (W.X.); (D.M.)
- Branch of National Clinical Research Center for Metabolic Diseases, Wuhan 430030, China
| | - Juan Chen
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China;
| | - Yan Yang
- Department of Endocrinology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (X.P.); (R.F.); (L.X.); (X.S.); (K.D.); (S.Z.); (J.T.); (W.X.); (D.M.)
- Branch of National Clinical Research Center for Metabolic Diseases, Wuhan 430030, China
| |
Collapse
|
36
|
Schütz L, Sixel-Döring F, Hermann W. Management of Sleep Disturbances in Parkinson's Disease. JOURNAL OF PARKINSON'S DISEASE 2022; 12:2029-2058. [PMID: 35938257 PMCID: PMC9661340 DOI: 10.3233/jpd-212749] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 06/23/2022] [Indexed: 06/07/2023]
Abstract
Parkinson's disease (PD) is defined by its motor symptoms rigidity, tremor, and akinesia. However, non-motor symptoms, particularly autonomic disorders and sleep disturbances, occur frequently in PD causing equivalent or even greater discomfort than motor symptoms effectively decreasing quality of life in patients and caregivers. Most common sleep disturbances in PD are insomnia, sleep disordered breathing, excessive daytime sleepiness, REM sleep behavior disorder, and sleep-related movement disorders such as restless legs syndrome. Despite their high prevalence, therapeutic options in the in- and outpatient setting are limited, partly due to lack of scientific evidence. The importance of sleep disturbances in neurodegenerative diseases has been further emphasized by recent evidence indicating a bidirectional relationship between neurodegeneration and sleep. A more profound insight into the underlying pathophysiological mechanisms intertwining sleep and neurodegeneration might lead to unique and individually tailored disease modifying or even neuroprotective therapeutic options in the long run. Therefore, current evidence concerning the management of sleep disturbances in PD will be discussed with the aim of providing a substantiated scaffolding for clinical decisions in long-term PD therapy.
Collapse
Affiliation(s)
- Lukas Schütz
- Department of Neurology, University of Rostock, Rostock, Germany
| | | | - Wiebke Hermann
- Department of Neurology, University of Rostock, Rostock, Germany
| |
Collapse
|
37
|
Won E, Na KS, Kim YK. Associations between Melatonin, Neuroinflammation, and Brain Alterations in Depression. Int J Mol Sci 2021; 23:ijms23010305. [PMID: 35008730 PMCID: PMC8745430 DOI: 10.3390/ijms23010305] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 12/21/2021] [Accepted: 12/26/2021] [Indexed: 12/14/2022] Open
Abstract
Pro-inflammatory systemic conditions that can cause neuroinflammation and subsequent alterations in brain regions involved in emotional regulation have been suggested as an underlying mechanism for the pathophysiology of major depressive disorder (MDD). A prominent feature of MDD is disruption of circadian rhythms, of which melatonin is considered a key moderator, and alterations in the melatonin system have been implicated in MDD. Melatonin is involved in immune system regulation and has been shown to possess anti-inflammatory properties in inflammatory conditions, through both immunological and non-immunological actions. Melatonin has been suggested as a highly cytoprotective and neuroprotective substance and shown to stimulate all stages of neuroplasticity in animal models. The ability of melatonin to suppress inflammatory responses through immunological and non-immunological actions, thus influencing neuroinflammation and neurotoxicity, along with subsequent alterations in brain regions that are implicated in depression, can be demonstrated by the antidepressant-like effects of melatonin. Further studies that investigate the associations between melatonin, immune markers, and alterations in the brain structure and function in patients with depression could identify potential MDD biomarkers.
Collapse
Affiliation(s)
- Eunsoo Won
- Department of Psychiatry, Chaum, Seoul 06062, Korea;
- Department of Psychiatry, CHA Bundang Medical Center, CHA University, Seongnam 13496, Korea
| | - Kyoung-Sae Na
- Department of Psychiatry, Gachon University Gil Medical Center, Incheon 21565, Korea;
| | - Yong-Ku Kim
- Department of Psychiatry, Korea University Ansan Hospital, Korea University College of Medicine, Ansan 15355, Korea
- Correspondence:
| |
Collapse
|
38
|
Devenney EM, McErlean K, Tse NY, Caga J, Dharmadasa T, Huynh W, Mahoney CJ, Zoing M, Mazumder S, Dobson-Stone C, Kwok JB, Halliday GM, Hodges JR, Piguet O, Ahmed RM, Kiernan MC. Factors That Influence Non-Motor Impairment Across the ALS-FTD Spectrum: Impact of Phenotype, Sex, Age, Onset and Disease Stage. Front Neurol 2021; 12:743688. [PMID: 34899567 PMCID: PMC8656429 DOI: 10.3389/fneur.2021.743688] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Accepted: 10/26/2021] [Indexed: 11/13/2022] Open
Abstract
Objective: This study aimed to establish (1) the pattern and severity of neuropsychiatric symptoms and other non-motor symptoms of sleep and mood, across ALS phenotypes in comparison to bvFTD and (2) the contribution of non-modifiable factors including age, sex and disease state to the severity of symptoms experienced by ALS patients. Methods: Consecutive participants were recruited to the study and underwent a detailed clinical, cognitive, behavioral and neuroimaging assessment. Neuropsychiatric and other non-motor symptoms were determined using the Cambridge Behavioral Inventory, the CBI-R. The scores were converted to define impairment in terms of mild, moderate and severe symptoms for each subscale. Rate, severity and contribution of King's staging and modifiable factors were also determined and a regression model identified predictors of symptom severity. Results: In total, 250 participants (115 ALS, 98 bvFTD, and 37 ALS-FTD patients) were recruited. A similar pattern of neuropsychiatric symptom severity was identified (apathy, disinhibition and stereotypic behavior) for all behavioral phenotypes of ALS compared to bvFTD (all p > 0.05). Neuropsychiatric symptoms were also present in cases defined as ALSpure and the cognitive phenotype of ALS (ALSci) although they occurred less frequently and were at the milder end of the spectrum. Disordered sleep and disrupted mood were common across all phenotypes (all p < 0.05). The severity of sleep dysfunction was influenced by both sex and age (all p < 0.05). Neuropsychiatric symptoms, sleep and mood disorders were common early in the disease process and deteriorated in line with progression on the Amyotrophic Lateral Sclerosis Functional Rating Scale-Revised (ALSFRS-R; all p < 0.05). Diagnostic phenotype, disease duration and global cognition scores were the strongest predictors of non-motor and neuropsychiatric impairments. Conclusion: The current findings reveal strikingly similar patterns of changes across the subgroups of ALS and bvFTD, supporting the concept of the ALS-FTD spectrum. The findings further highlight the impact of non-motor and neuropsychiatric symptoms in patients with ALS, that are often as severe as that seen in ALS-FTD and bvFTD. This study advances understanding across the ALS-FTD spectrum that may accelerate the early identification of patient needs, to ensure prompt recognition of symptoms and thereby to improve clinical awareness, patient care and management.
Collapse
Affiliation(s)
- Emma M Devenney
- Brain and Mind Centre, The University of Sydney, Sydney, NSW, Australia.,Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| | - Kate McErlean
- Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Nga Yan Tse
- Brain and Mind Centre, The University of Sydney, Sydney, NSW, Australia.,Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| | - Jashelle Caga
- Brain and Mind Centre, The University of Sydney, Sydney, NSW, Australia.,Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| | - Thanuja Dharmadasa
- Brain and Mind Centre, The University of Sydney, Sydney, NSW, Australia.,Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia.,Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, United Kingdom
| | - William Huynh
- Brain and Mind Centre, The University of Sydney, Sydney, NSW, Australia.,Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia.,Prince of Wales Clinical School, University of New South Wales, Kensington, NSW, Australia
| | - Colin J Mahoney
- Brain and Mind Centre, The University of Sydney, Sydney, NSW, Australia.,Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| | - Margaret Zoing
- Brain and Mind Centre, The University of Sydney, Sydney, NSW, Australia.,Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| | - Srestha Mazumder
- Brain and Mind Centre, The University of Sydney, Sydney, NSW, Australia.,Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| | - Carol Dobson-Stone
- Brain and Mind Centre, The University of Sydney, Sydney, NSW, Australia.,Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| | - John B Kwok
- Brain and Mind Centre, The University of Sydney, Sydney, NSW, Australia.,Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| | - Glenda M Halliday
- Brain and Mind Centre, The University of Sydney, Sydney, NSW, Australia.,Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| | - John R Hodges
- Brain and Mind Centre, The University of Sydney, Sydney, NSW, Australia.,Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| | - Olivier Piguet
- Brain and Mind Centre, The University of Sydney, Sydney, NSW, Australia.,School of Psychology, The University of Sydney, Sydney, NSW, Australia
| | - Rebekah M Ahmed
- Brain and Mind Centre, The University of Sydney, Sydney, NSW, Australia.,Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia.,Institute of Clinical Neurosciences, Royal Prince Alfred Hospital, Camperdown, NSW, Australia
| | - Matthew C Kiernan
- Brain and Mind Centre, The University of Sydney, Sydney, NSW, Australia.,Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia.,Institute of Clinical Neurosciences, Royal Prince Alfred Hospital, Camperdown, NSW, Australia
| |
Collapse
|
39
|
Ahn Y, Han SH, Kim MG, Hong KB, Kim WJ, Suh HJ, Jo K. Anti-depressant effects of ethanol extract from Cannabis sativa (hemp) seed in chlorpromazine-induced Drosophila melanogaster depression model. PHARMACEUTICAL BIOLOGY 2021; 59:998-1007. [PMID: 34362287 PMCID: PMC8354181 DOI: 10.1080/13880209.2021.1949356] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/15/2023]
Abstract
CONTEXT Depression is a severe mental illness caused by a deficiency of dopamine and serotonin. Cannabis sativa L. (Cannabaceae) has long been used to treat pain, nausea, and depression. OBJECTIVE This study investigates the anti-depressant effects of C. sativa (hemp) seed ethanol extract (HE) in chlorpromazine (CPZ)-induced Drosophila melanogaster depression model. MATERIALS AND METHODS The normal group was untreated, and the control group was treated with CPZ (0.1% of media) for 7 days. The experimental groups were treated with a single HE treatment (0.5, 1.0, and 1.5% of media) and a mixture of 0.1% CPZ and HE for 7 days. The locomotor activity, behavioural patterns, depression-related gene expression, and neurotransmitters level of flies were investigated. RESULTS The behavioural patterns of individual flies were significantly reduced with 0.1% CPZ treatment. In contrast, combination treatment of 1.5% HE and 0.1% CPZ significantly increased subjective daytime activity (p < 0.001) and behavioural factors (p < 0.001). These results correlate with increased transcript levels of dopamine (p < 0.001) and serotonin (p < 0.05) receptors and concentration of dopamine (p < 0.05), levodopa (p < 0.001), 5-HTP (p < 0.05), and serotonin (p < 0.001) compared to those in the control group. DISCUSSION AND CONCLUSIONS Collectively, HE administration alleviates depression-like symptoms by modulating the circadian rhythm-related behaviours, transcript levels of neurotransmitter receptors, and neurotransmitter levels in the CPZ-induced Drosophila model. However, additional research is needed to investigate the role of HE administration in behavioural patterns, reduction of the neurotransmitter, and signalling pathways of depression in a vertebrate model system.
Collapse
Affiliation(s)
- Yejin Ahn
- Department of Integrated Biomedical and Life Science, Graduate School, Korea University, Seoul, Republic of Korea
| | - Sung Hee Han
- Institute of Human Behavior & Genetic, College of Medicine, Korea University, Seoul, Republic of Korea
| | - Min Guk Kim
- Department of Integrated Biomedical and Life Science, Graduate School, Korea University, Seoul, Republic of Korea
| | - Ki-Bae Hong
- Department of Food Science and Nutrition, Jeju National University, Jeju, Republic of Korea
| | - Woo Jung Kim
- Biocenter, Gyeonggido Business and Science Accerlerator, Suwon, Republic of Korea
| | - Hyung Joo Suh
- Department of Integrated Biomedical and Life Science, Graduate School, Korea University, Seoul, Republic of Korea
| | - Kyungae Jo
- Department of Integrated Biomedical and Life Science, Graduate School, Korea University, Seoul, Republic of Korea
- CONTACT Kyungae Jo Department of Integrated Biomedical and Life Science, Graduate School, Korea University, Seoul, Republic of Korea
| |
Collapse
|
40
|
Niu L, Zhang F, Xu X, Yang Y, Li S, Liu H, Le W. Chronic sleep deprivation altered the expression of circadian clock genes and aggravated Alzheimer's disease neuropathology. Brain Pathol 2021; 32:e13028. [PMID: 34668266 PMCID: PMC9048513 DOI: 10.1111/bpa.13028] [Citation(s) in RCA: 66] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2021] [Revised: 09/18/2021] [Accepted: 09/27/2021] [Indexed: 01/20/2023] Open
Abstract
Circadian disruption is prevalent in Alzheimer's disease (AD) and may contribute to cognitive impairment, psychological symptoms, and neurodegeneration. This study aimed to evaluate the effects of environmental and genetic factors on the molecular clock and to establish a link between circadian rhythm disturbance and AD. We investigated the pathological effects of chronic sleep deprivation (CSD) in the APPswe/PS1ΔE9 transgenic mice and their wild‐type (WT) littermates for 2 months and evaluated the expression levels of clock genes in the circadian rhythm‐related nuclei. Our results showed that CSD impaired learning and memory, and further exaggerated disease progression in the AD mice. Furthermore, CSD caused abnormal expression of Bmal1, Clock, and Cry1 in the circadian rhythm‐related nuclei of experimental mice, and these changes are more significant in AD mice. Abnormal expression of clock genes in AD mice suggested that the expression of clock genes is affected by APP/PS1 mutations. In addition, abnormal tau phosphorylation was found in the retrosplenial cortex, which was co‐located with the alteration of BMAL1 protein level. Moreover, the level of tyrosine hydroxylase in the locus coeruleus of AD and WT mice was significantly increased after CSD. There may be a potential link between the molecular clock, Aβ pathology, tauopathy, and the noradrenergic system. The results of this study provided new insights into the potential link between the disruption of circadian rhythm and the development of AD.
Collapse
Affiliation(s)
- Long Niu
- Center for Clinical Research on Neurological Diseases, the First Affiliated Hospital, Dalian Medical University, Dalian, China.,Liaoning Provincial Key Laboratory for Research on the Pathogenic Mechanisms of Neurological Diseases, the First Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Feng Zhang
- Center for Clinical Research on Neurological Diseases, the First Affiliated Hospital, Dalian Medical University, Dalian, China.,Liaoning Provincial Key Laboratory for Research on the Pathogenic Mechanisms of Neurological Diseases, the First Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Xiaojiao Xu
- Center for Clinical Research on Neurological Diseases, the First Affiliated Hospital, Dalian Medical University, Dalian, China.,Liaoning Provincial Key Laboratory for Research on the Pathogenic Mechanisms of Neurological Diseases, the First Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Yuting Yang
- Center for Clinical Research on Neurological Diseases, the First Affiliated Hospital, Dalian Medical University, Dalian, China.,Liaoning Provincial Key Laboratory for Research on the Pathogenic Mechanisms of Neurological Diseases, the First Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Song Li
- Center for Clinical Research on Neurological Diseases, the First Affiliated Hospital, Dalian Medical University, Dalian, China.,Liaoning Provincial Key Laboratory for Research on the Pathogenic Mechanisms of Neurological Diseases, the First Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Hui Liu
- Department of Neurology, Minhang Hospital, Fudan University, Shanghai, China
| | - Weidong Le
- Center for Clinical Research on Neurological Diseases, the First Affiliated Hospital, Dalian Medical University, Dalian, China.,Liaoning Provincial Key Laboratory for Research on the Pathogenic Mechanisms of Neurological Diseases, the First Affiliated Hospital, Dalian Medical University, Dalian, China.,Department of Neurology and Institute of Neurology, Sichuan Academy of Medical Science-Sichuan Provincial Hospital, Chengdu, China
| |
Collapse
|
41
|
Maiese K. Neurodegeneration, memory loss, and dementia: the impact of biological clocks and circadian rhythm. FRONT BIOSCI-LANDMRK 2021; 26:614-627. [PMID: 34590471 PMCID: PMC8756734 DOI: 10.52586/4971] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2021] [Revised: 07/26/2021] [Accepted: 08/10/2021] [Indexed: 11/23/2022]
Abstract
Introduction: Dementia and cognitive loss impact a significant proportion of the global population and present almost insurmountable challenges for treatment since they stem from multifactorial etiologies. Innovative avenues for treatment are highly warranted. Methods and results: Novel work with biological clock genes that oversee circadian rhythm may meet this critical need by focusing upon the pathways of the mechanistic target of rapamycin (mTOR), the silent mating type information regulation 2 homolog 1 (Saccharomyces cerevisiae) (SIRT1), mammalian forkhead transcription factors (FoxOs), the growth factor erythropoietin (EPO), and the wingless Wnt pathway. These pathways are complex in nature, intimately associated with autophagy that can maintain circadian rhythm, and have an intricate relationship that can lead to beneficial outcomes that may offer neuroprotection, metabolic homeostasis, and prevention of cognitive loss. However, biological clocks and alterations in circadian rhythm also have the potential to lead to devastating effects involving tumorigenesis in conjunction with pathways involving Wnt that oversee angiogenesis and stem cell proliferation. Conclusions: Current work with biological clocks and circadian rhythm pathways provide exciting possibilities for the treating dementia and cognitive loss, but also provide powerful arguments to further comprehend the intimate and complex relationship among these pathways to fully potentiate desired clinical outcomes.
Collapse
Affiliation(s)
- Kenneth Maiese
- Cellular and Molecular Signaling, New York, NY 10022, USA
| |
Collapse
|
42
|
Wang XL, Li L. Circadian Clock Regulates Inflammation and the Development of Neurodegeneration. Front Cell Infect Microbiol 2021; 11:696554. [PMID: 34595127 PMCID: PMC8476957 DOI: 10.3389/fcimb.2021.696554] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Accepted: 08/18/2021] [Indexed: 12/15/2022] Open
Abstract
The circadian clock regulates numerous key physiological processes and maintains cellular, tissue, and systemic homeostasis. Disruption of circadian clock machinery influences key activities involved in immune response and brain function. Moreover, Immune activation has been closely linked to neurodegeneration. Here, we review the molecular clock machinery and the diurnal variation of immune activity. We summarize the circadian control of immunity in both central and peripheral immune cells, as well as the circadian regulation of brain cells that are implicated in neurodegeneration. We explore the important role of systemic inflammation on neurodegeneration. The circadian clock modulates cellular metabolism, which could be a mechanism underlying circadian control. We also discuss the circadian interventions implicated in inflammation and neurodegeneration. Targeting circadian clocks could be a potential strategy for the prevention and treatment of inflammation and neurodegenerative diseases.
Collapse
Affiliation(s)
- Xiao-Lan Wang
- Department of Nephrology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lianjian Li
- Department of Surgery, Hubei Provincial Hospital of Traditional Chinese Medicine, Wuhan, China
- Hubei Province Academy of Traditional Chinese Medicine, Wuhan, China
| |
Collapse
|
43
|
Maiese K. Cognitive Impairment and Dementia: Gaining Insight through Circadian Clock Gene Pathways. Biomolecules 2021; 11:1002. [PMID: 34356626 PMCID: PMC8301848 DOI: 10.3390/biom11071002] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 07/05/2021] [Accepted: 07/07/2021] [Indexed: 01/18/2023] Open
Abstract
Neurodegenerative disorders affect fifteen percent of the world's population and pose a significant financial burden to all nations. Cognitive impairment is the seventh leading cause of death throughout the globe. Given the enormous challenges to treat cognitive disorders, such as Alzheimer's disease, and the inability to markedly limit disease progression, circadian clock gene pathways offer an exciting strategy to address cognitive loss. Alterations in circadian clock genes can result in age-related motor deficits, affect treatment regimens with neurodegenerative disorders, and lead to the onset and progression of dementia. Interestingly, circadian pathways hold an intricate relationship with autophagy, the mechanistic target of rapamycin (mTOR), the silent mating type information regulation 2 homolog 1 (Saccharomyces cerevisiae) (SIRT1), mammalian forkhead transcription factors (FoxOs), and the trophic factor erythropoietin. Autophagy induction is necessary to maintain circadian rhythm homeostasis and limit cortical neurodegenerative disease, but requires a fine balance in biological activity to foster proper circadian clock gene regulation that is intimately dependent upon mTOR, SIRT1, FoxOs, and growth factor expression. Circadian rhythm mechanisms offer innovative prospects for the development of new avenues to comprehend the underlying mechanisms of cognitive loss and forge ahead with new therapeutics for dementia that can offer effective clinical treatments.
Collapse
Affiliation(s)
- Kenneth Maiese
- Cellular and Molecular Signaling, New York, NY 10022, USA
| |
Collapse
|
44
|
Liu Y, Niu L, Liu X, Cheng C, Le W. Recent Progress in Non-motor Features of Parkinson's Disease with a Focus on Circadian Rhythm Dysregulation. Neurosci Bull 2021; 37:1010-1024. [PMID: 34128188 DOI: 10.1007/s12264-021-00711-x] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Accepted: 03/31/2021] [Indexed: 12/14/2022] Open
Abstract
Parkinson's disease (PD) is the second most common neurodegenerative disease, which manifests with both motor and non-motor symptoms. Circadian rhythm dysregulation, as one of the most challenging non-motor features of PD, usually appears long before obvious motor symptoms. Moreover, the dysregulated circadian rhythm has recently been reported to play pivotal roles in PD pathogenesis, and it has emerged as a hot topic in PD research. In this review, we briefly introduce the circadian rhythm and circadian rhythm-related genes, and then summarize recent research progress on the altered circadian rhythm in PD, ranging from clinical features to the possible causes of PD-related circadian disorders. We believe that future comprehensive studies on the topic may not only help us to explore the mechanisms of PD, but also shed light on the better management of PD.
Collapse
Affiliation(s)
- Yufei Liu
- Liaoning Provincial Key Laboratory for Research on the Pathogenic Mechanisms of Neurological Diseases, the First Affiliated Hospital, Dalian Medical University, Dalian, 116021, China
- Liaoning Provincial Center for Clinical Research on Neurological Diseases, the First Affiliated Hospital, Dalian Medical University, Dalian, 116021, China
| | - Long Niu
- Liaoning Provincial Key Laboratory for Research on the Pathogenic Mechanisms of Neurological Diseases, the First Affiliated Hospital, Dalian Medical University, Dalian, 116021, China
- Liaoning Provincial Center for Clinical Research on Neurological Diseases, the First Affiliated Hospital, Dalian Medical University, Dalian, 116021, China
| | - Xinyao Liu
- Liaoning Provincial Key Laboratory for Research on the Pathogenic Mechanisms of Neurological Diseases, the First Affiliated Hospital, Dalian Medical University, Dalian, 116021, China
- Liaoning Provincial Center for Clinical Research on Neurological Diseases, the First Affiliated Hospital, Dalian Medical University, Dalian, 116021, China
| | - Cheng Cheng
- Liaoning Provincial Key Laboratory for Research on the Pathogenic Mechanisms of Neurological Diseases, the First Affiliated Hospital, Dalian Medical University, Dalian, 116021, China
- Liaoning Provincial Center for Clinical Research on Neurological Diseases, the First Affiliated Hospital, Dalian Medical University, Dalian, 116021, China
| | - Weidong Le
- Liaoning Provincial Key Laboratory for Research on the Pathogenic Mechanisms of Neurological Diseases, the First Affiliated Hospital, Dalian Medical University, Dalian, 116021, China.
- Liaoning Provincial Center for Clinical Research on Neurological Diseases, the First Affiliated Hospital, Dalian Medical University, Dalian, 116021, China.
- Institute of Neurology, Sichuan Academy of Medical Science-Sichuan Provincial Hospital, University of Electronic Science and Technology of China, Chengdu, 610072, China.
| |
Collapse
|
45
|
Sanchez REA, Kalume F, de la Iglesia HO. Sleep timing and the circadian clock in mammals: Past, present and the road ahead. Semin Cell Dev Biol 2021; 126:3-14. [PMID: 34092510 DOI: 10.1016/j.semcdb.2021.05.034] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 05/25/2021] [Accepted: 05/31/2021] [Indexed: 01/22/2023]
Abstract
Nearly all mammals display robust daily rhythms of physiology and behavior. These approximately 24-h cycles, known as circadian rhythms, are driven by a master clock in the suprachiasmatic nucleus (SCN) of the hypothalamus and affect biological processes ranging from metabolism to immune function. Perhaps the most overt output of the circadian clock is the sleep-wake cycle, the integrity of which is critical for health and homeostasis of the organism. In this review, we summarize our current understanding of the circadian regulation of sleep. We discuss the neural circuitry and molecular mechanisms underlying daily sleep timing, and the trajectory of circadian regulation of sleep across development. We conclude by proposing future research priorities for the field that will significantly advance our mechanistic understanding of the circadian regulation of sleep.
Collapse
Affiliation(s)
- Raymond E A Sanchez
- Department of Biology, University of Washington, Seattle, WA, USA; Graduate Program in Neuroscience, University of Washington, Seattle, WA, USA.
| | - Franck Kalume
- Graduate Program in Neuroscience, University of Washington, Seattle, WA, USA; Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA, USA; Department of Neurological Surgery, University of Washington, Seattle, WA, USA; Department of Pharmacology, University of Washington, Seattle, WA, USA
| | - Horacio O de la Iglesia
- Department of Biology, University of Washington, Seattle, WA, USA; Graduate Program in Neuroscience, University of Washington, Seattle, WA, USA
| |
Collapse
|
46
|
Vanderheyden WM, Fang B, Flores CC, Jager J, Gerstner JR. The transcriptional repressor Rev-erbα regulates circadian expression of the astrocyte Fabp7 mRNA. CURRENT RESEARCH IN NEUROBIOLOGY 2021; 2. [PMID: 34056625 PMCID: PMC8162199 DOI: 10.1016/j.crneur.2021.100009] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
The astrocyte brain-type fatty-acid binding protein (Fabp7) circadian gene expression is synchronized in the same temporal phase throughout mammalian brain. Cellular and molecular mechanisms that contribute to this coordinated expression are not completely understood, but likely involve the nuclear receptor Rev-erbα (NR1D1), a transcriptional repressor. We performed ChIP-seq on ventral tegmental area (VTA) and identified gene targets of Rev-erbα, including Fabp7. We confirmed that Rev-erbα binds to the Fabp7 promoter in multiple brain areas, including hippocampus, hypothalamus, and VTA, and showed that Fabp7 gene expression is upregulated in Rev-erbα knock-out mice. Compared to Fabp7 mRNA levels, Fabp3 and Fabp5 mRNA were unaffected by Rev-erbα depletion in hippocampus, suggesting that these effects are specific to Fabp7. To determine whether these effects of Rev-erbα depletion occur broadly throughout the brain, we also evaluated Fabp mRNA expression levels in multiple brain areas, including cerebellum, cortex, hypothalamus, striatum, and VTA in Rev-erbα knock-out mice. While small but significant changes in Fabp5 mRNA expression exist in some of these areas, the magnitude of these effects are minimal to that of Fabp7 mRNA expression, which was over 6-fold across all brain regions. These studies suggest that Rev-erbα is a transcriptional repressor of Fabp7 gene expression throughout mammalian brain. The transcriptional repressor Rev-erbα binds to the Fabp7 promoter across brain areas. Multiple Rev-erbα response element binding sites exist on the Fabp7 promoter. Rev-erbα is required for Fabp7 transcriptional repression and circadian expression. Rev-erbα depletion does not affect other Fabp-type gene expression in brain.
Collapse
Affiliation(s)
- William M Vanderheyden
- Elson S. Floyd College of Medicine, Washington State University, Spokane, WA. 99202, USA.,Sleep and Performance Research Center, Washington State University, Spokane, WA. 99202, USA
| | - Bin Fang
- Genomics Institute of the Novartis Research Foundation, 10675 John Jay Hopkins Dr, San Diego, CA 92121
| | - Carlos C Flores
- Elson S. Floyd College of Medicine, Washington State University, Spokane, WA. 99202, USA
| | - Jennifer Jager
- Université Côte d'Azur, INSERM, Centre Méditerranéen de Médecine Moléculaire (C3M), Cellular and Molecular Physiopathology of Obesity, Nice, France
| | - Jason R Gerstner
- Elson S. Floyd College of Medicine, Washington State University, Spokane, WA. 99202, USA.,Sleep and Performance Research Center, Washington State University, Spokane, WA. 99202, USA.,Steve Gleason Institute for Neuroscience, Washington State University, Spokane, WA. 99202, USA
| |
Collapse
|
47
|
Khayachi A, Schorova L, Alda M, Rouleau GA, Milnerwood AJ. Posttranslational modifications & lithium's therapeutic effect-Potential biomarkers for clinical responses in psychiatric & neurodegenerative disorders. Neurosci Biobehav Rev 2021; 127:424-445. [PMID: 33971223 DOI: 10.1016/j.neubiorev.2021.05.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Revised: 03/14/2021] [Accepted: 05/03/2021] [Indexed: 01/03/2023]
Abstract
Several neurodegenerative diseases and neuropsychiatric disorders display aberrant posttranslational modifications (PTMs) of one, or many, proteins. Lithium treatment has been used for mood stabilization for many decades, and is highly effective for large subsets of patients with diverse neurological conditions. However, the differential effectiveness and mode of action are not fully understood. In recent years, studies have shown that lithium alters several protein PTMs, altering their function, and consequently neuronal physiology. The impetus for this review is to outline the links between lithium's therapeutic mode of action and PTM homeostasis. We first provide an overview of the principal PTMs affected by lithium. We then describe several neuropsychiatric disorders in which PTMs have been implicated as pathogenic. For each of these conditions, we discuss lithium's clinical use and explore the putative mechanism of how it restores PTM homeostasis, and thereby cellular physiology. Evidence suggests that determining specific PTM patterns could be a promising strategy to develop biomarkers for disease and lithium responsiveness.
Collapse
Affiliation(s)
- A Khayachi
- Montreal Neurological Institute, Department of Neurology & Neurosurgery, McGill University, Montréal, Quebec, Canada.
| | - L Schorova
- McGill University Health Center Research Institute, Montréal, Quebec, Canada
| | - M Alda
- Department of Psychiatry, Dalhousie University, Halifax, Nova Scotia, Canada
| | - G A Rouleau
- Montreal Neurological Institute, Department of Neurology & Neurosurgery, McGill University, Montréal, Quebec, Canada; Department of Human Genetics, McGill University, Montréal, Quebec, Canada.
| | - A J Milnerwood
- Montreal Neurological Institute, Department of Neurology & Neurosurgery, McGill University, Montréal, Quebec, Canada.
| |
Collapse
|
48
|
Mayer G, Happe S, Evers S, Hermann W, Jansen S, Kallweit U, Muntean ML, Pöhlau D, Riemann D, Saletu M, Schichl M, Schmitt WJ, Sixel-Döring F, Young P. Insomnia in neurological diseases. Neurol Res Pract 2021; 3:15. [PMID: 33691803 PMCID: PMC7944611 DOI: 10.1186/s42466-021-00106-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2021] [Accepted: 01/12/2021] [Indexed: 11/13/2022] Open
Abstract
Insomnia is defined as difficulties of initiating and maintaining sleep, early awakening and poor subjective sleep quality despite adequate opportunity and circumstances for sleep with impairment of daytime performance. These components of insomnia - namely persistent sleep difficulties despite of adequate sleep opportunity resulting in daytime dysfunction - appear secondary or co-morbid to neurological diseases. Comorbid insomnia originates from neurodegenerative, inflammatory, traumatic or ischemic changes in sleep regulating brainstem and hypothalamic nuclei with consecutive changes of neurotransmitters. Symptoms of neurological disorders (i.e motor deficits), co-morbidities (i.e. pain, depression, anxiety) and some disease-specific pharmaceuticals may cause insomnia and/or other sleep problems.This guideline focuses on insomnias in headaches, neurodegenerative movement disorders, multiple sclerosis, traumatic brain injury, epilepsies, stroke, neuromuscular disease and dementia.The most important new recommendations are: Cognitive behavioral therapy (CBTi) is recommended to treat acute and chronic insomnia in headache patients. Insomnia is one of the most frequent sleep complaints in neurodegenerative movement disorders. Patients may benefit from CBTi, antidepressants (trazodone, doxepin), melatonin and gaba-agonists. Insomnia is a frequent precursor of MS symptoms by up to 10 years. CBTi is recommended in patients with MS, traumatic brain injury and. Melatonin may improve insomnia symptoms in children with epilepsies. Patients with insomnia after stroke can be treated with benzodiazepine receptor agonists and sedating antidepressants. For patients with dementia suffering from insomnia trazodone, light therapy and physical exercise are recommended.
Collapse
Affiliation(s)
- Geert Mayer
- Neurologische Abteilung der Hephata-Klinik, Schimmelpfengstrasse 6, 34613, Schwalmstadt-Treysa, Germany.
- Neurologische Abteilung der Philipps-Universität Marburg, Mamburg, Germany.
| | - Svenja Happe
- Klinik Maria Frieden, Klinik für Neurologie, Am Krankenhaus 1, 48291, Telgte, Germany
| | - Stefan Evers
- Krankenhaus Lindenbrunn, Abteilung Neurologie, Lindenbrunn 1, 31863, Coppenbrügge, Germany
| | - Wiebke Hermann
- Klinik und Poliklinik für Neurologie und Deutsches Zentrum für Neurodegenerative Erkrankungen e.V. (DZNE), Gehlsheimer Str. 20, 18147, Rostock, Germany
| | - Sabine Jansen
- Deutsche Alzheimer Gesellschaft e.V. Selbsthilfe Demenz, Friedrichstr. 236, 10969, Berlin, Germany
| | - Ulf Kallweit
- Klin. Schlaf- und Neuroimmunologie, Private Universität Witten/Herdecke gGmbH, Alfred-Herrhausen-Str. 50, 58448, Witten, Germany
| | - Maria-Lucia Muntean
- Paracelsus Elena Klinik, Schanzenstr. 85 Dr. med Dieter Pöhlau, 34130, Kassel, Germany
- DRK Kamillus Klinik, Hospitalstr. 6, 53567, Asbach, Germany
| | - Dieter Pöhlau
- DRK Kamillus Klinik, Hospitalstr. 6, 53567, Asbach, Germany
| | - Dieter Riemann
- Psychiatrische Universitätsklinik Freiburg, Hauptstraße 5, 79104, Freiburg, Germany
| | - Michael Saletu
- LKH - Graz II, Standort Süd, Wagner Jauregg Platz 1, A-8053, Graz, Austria
| | | | - Wolfgang J Schmitt
- Universitätsklinik für Psychiatrie und Psychotherapie, Murtenstrasse 21, 3008, Bern, Switzerland
| | | | - Peter Young
- Neurologische Klinik Reithofpark, Reithof 1, 83075, Bad Feilnbach, Germany
| |
Collapse
|
49
|
Yang Z, Zhang X, Li C, Chi S, Xie A. Molecular Mechanisms Underlying Reciprocal Interactions Between Sleep Disorders and Parkinson's Disease. Front Neurosci 2021; 14:592989. [PMID: 33642969 PMCID: PMC7902929 DOI: 10.3389/fnins.2020.592989] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2020] [Accepted: 11/27/2020] [Indexed: 01/11/2023] Open
Abstract
Sleep-wake disruptions are among the most prevalent and burdensome non-motor symptoms of Parkinson's disease (PD). Clinical studies have demonstrated that these disturbances can precede the onset of typical motor symptoms by years, indicating that they may play a primary function in the pathogenesis of PD. Animal studies suggest that sleep facilitates the removal of metabolic wastes through the glymphatic system via convective flow from the periarterial space to the perivenous space, upregulates antioxidative defenses, and promotes the maintenance of neuronal protein homeostasis. Therefore, disruptions to the sleep-wake cycle have been associated with inefficient metabolic clearance and increased oxidative stress in the central nervous system (CNS). This leads to excessive accumulation of alpha-synuclein and the induction of neuronal loss, both of which have been proposed to be contributing factors to the pathogenesis and progression of PD. Additionally, recent studies have suggested that PD-related pathophysiological alterations during the prodromal phase disrupt sleep and circadian rhythms. Taken together, these findings indicate potential mechanistic interactions between sleep-wake disorders and PD progression as proposed in this review. Further research into the hypothetical mechanisms underlying these interactions would be valuable, as positive findings may provide promising insights into novel therapeutic interventions for PD.
Collapse
Affiliation(s)
- Zhengjie Yang
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Xiaona Zhang
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Chengqian Li
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Song Chi
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Anmu Xie
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao, China
| |
Collapse
|
50
|
Khyati, Malik I, Agrawal N, Kumar V. Melatonin and curcumin reestablish disturbed circadian gene expressions and restore locomotion ability and eclosion behavior in Drosophila model of Huntington's disease. Chronobiol Int 2020; 38:61-78. [PMID: 33334207 DOI: 10.1080/07420528.2020.1842752] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Deficit in locomotion (motor) ability and disturbance of the circadian behavior and sleep-wake pattern characterize Huntington's disease (HD). Here, we examined the disturbance of circadian timing with the progression of HD pathogenesis, and tested the efficacy of melatonin and curcumin in preventing the motor deficit and disturbed eclosion behavior in the Drosophila model of HD. To examine circadian timing, we assayed mRNA expression of genes of the transcriptional feedback (TF) loop that generates the near 24-h rhythmicity. We performed qPCR of the Period, Timeless, Clock, Cycle, Clockwork, and Cryptochrome genes in transgenic fly heads from elav-Gal4 (pan neuronal) and PDF-Gal4 (PDF-specific neurons) driver lines through the progression of HD disease post-eclosion, from day 1 to its terminal stage on day 13. Cycle was arrhythmic from day 1, but Period and Timeless became arrhythmic on day 13 of the HD pathogenesis in elav, but not PDF, neurons. Twenty-four-hour mRNA rhythms showed alteration in the waveform properties (mesor and amplitude, not acrophase), but not in the persistence, in both elav-Gal4 and PDF-Gal4 HD flies; however, disturbance of the clock gene rhythm was delayed in PDF-Gal4 flies. To assess the preventive effects on HD pathogenesis, flies of both driver lines were provided with melatonin (50, 100, or 150 μg) or curcumin (10 μM) in the diet commencing from the larval stage. Both melatonin (100 μg) and curcumin reestablished the 24-h pattern in mRNA expression of Period and Timeless to normal (control) levels, and significantly improved both locomotion ability and eclosion behavior of HD flies. We suggest that the disturbance of circadian timekeeping progressively accelerated HD pathogenesis, possibly via modulation of the transcriptional state that resulted in the modification of the Huntington gene. These findings suggest melatonin and curcumin might be potential therapeutic agents for the treatment of HD in humans, although this needs specific investigation.
Collapse
Affiliation(s)
- Khyati
- Department of Zoology, University of Delhi , Delhi, India
| | - Indu Malik
- Department of Zoology, University of Delhi , Delhi, India
| | - Namita Agrawal
- Department of Zoology, University of Delhi , Delhi, India
| | - Vinod Kumar
- Department of Zoology, University of Delhi , Delhi, India
| |
Collapse
|